首页 > 最新文献

Progress in Tumor Research最新文献

英文 中文
Immunotherapy of Breast Cancer. 乳腺癌的免疫治疗。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2015-01-01 Epub Date: 2015-09-04 DOI: 10.1159/000437183
Carmen Criscitiello, Giuseppe Curigliano

Cancer immunoediting is the process by which the immune system protects the host from tumor development and guides the somatic evolution of tumors by eliminating highly immunogenic tumor cells. A fundamental dogma of tumor immunology and of cancer immunosurveillance in particular is that cancer cells express antigens that differentiate them from their nontransformed counterparts. Molecular studies clearly show that these antigens were often products of mutated cellular genes, aberrantly expressed normal genes, or genes encoding viral proteins. There is a strict correlation between genetic instability and the immune landscape of a breast cancer. Mutational heterogeneity in breast cancer is associated with new cancer-associated genes and new cancer antigens. Frequencies of somatic mutations or mutational burden can be related to the immunogenicity of breast cancer. We believe that molecular subtypes of breast cancer that are triple negative, luminal B-like or HER2-positive have a high mutational burden and can be considered immunogenic. The increasing knowledge of the immune system's capacity to not only recognize and destroy cancer, but also to shape cancer immunogenicity will develop more informed attempts to control cancer via immunological approaches. To be effective in breast cancer, immunotherapies will have to increase the quality or quantity of immune effector cells, reveal additional protective tumor antigens, and/or eliminate cancer-induced immunosuppressive mechanisms. Multiple immunotherapy approaches are under investigation in patients with breast cancer. These include vaccine approaches to elicit strong specific immune responses to tumor antigens such as WT-1, HER2 and NY-ESO-1, approaches involving adoptive transfer of in vitro-expanded, naturally arising or genetically engineered tumor-specific lymphocytes, therapeutic administration of monoclonal antibodies to target and eliminate tumor cells, and approaches that inhibit or destroy the molecular or cellular mediators of cancer-induced immunosuppression, such as CTLA-4, PD-1 or Treg cells. Here we provide a concise and comprehensive review on the role and utility of promising immunotherapeutics for the treatment of patients with breast cancer.

癌症免疫编辑是免疫系统通过消除高免疫原性肿瘤细胞来保护宿主免受肿瘤发展并指导肿瘤体细胞进化的过程。肿瘤免疫学,特别是癌症免疫监测的一个基本原则是,癌细胞表达抗原,使它们与未转化的对应细胞区分开来。分子研究清楚地表明,这些抗原通常是突变的细胞基因、异常表达的正常基因或编码病毒蛋白的基因的产物。遗传不稳定性和乳腺癌的免疫状况之间有着密切的联系。乳腺癌的突变异质性与新的癌症相关基因和新的癌症抗原有关。体细胞突变或突变负担的频率可能与乳腺癌的免疫原性有关。我们认为,三阴性、腔内b样或her2阳性的乳腺癌分子亚型具有很高的突变负担,可以认为是免疫原性的。随着人们对免疫系统不仅能识别和摧毁癌症,还能塑造癌症免疫原性的能力的认识不断增加,人们将开发出更明智的尝试,通过免疫方法控制癌症。为了对乳腺癌有效,免疫疗法必须提高免疫效应细胞的质量或数量,揭示额外的保护性肿瘤抗原,和/或消除癌症诱导的免疫抑制机制。对乳腺癌患者的多种免疫治疗方法正在研究中。这些方法包括对肿瘤抗原(如WT-1、HER2和NY-ESO-1)引发强烈特异性免疫反应的疫苗方法,涉及体外扩增、自然产生或基因工程的肿瘤特异性淋巴细胞过继转移的方法,靶向和消除肿瘤细胞的单克隆抗体治疗性管理,以及抑制或破坏癌症诱导免疫抑制的分子或细胞介质(如CTLA-4、PD-1或Treg细胞)的方法。在这里,我们提供了一个简明而全面的回顾的作用和有前途的免疫疗法的应用,以治疗乳腺癌患者。
{"title":"Immunotherapy of Breast Cancer.","authors":"Carmen Criscitiello,&nbsp;Giuseppe Curigliano","doi":"10.1159/000437183","DOIUrl":"https://doi.org/10.1159/000437183","url":null,"abstract":"<p><p>Cancer immunoediting is the process by which the immune system protects the host from tumor development and guides the somatic evolution of tumors by eliminating highly immunogenic tumor cells. A fundamental dogma of tumor immunology and of cancer immunosurveillance in particular is that cancer cells express antigens that differentiate them from their nontransformed counterparts. Molecular studies clearly show that these antigens were often products of mutated cellular genes, aberrantly expressed normal genes, or genes encoding viral proteins. There is a strict correlation between genetic instability and the immune landscape of a breast cancer. Mutational heterogeneity in breast cancer is associated with new cancer-associated genes and new cancer antigens. Frequencies of somatic mutations or mutational burden can be related to the immunogenicity of breast cancer. We believe that molecular subtypes of breast cancer that are triple negative, luminal B-like or HER2-positive have a high mutational burden and can be considered immunogenic. The increasing knowledge of the immune system's capacity to not only recognize and destroy cancer, but also to shape cancer immunogenicity will develop more informed attempts to control cancer via immunological approaches. To be effective in breast cancer, immunotherapies will have to increase the quality or quantity of immune effector cells, reveal additional protective tumor antigens, and/or eliminate cancer-induced immunosuppressive mechanisms. Multiple immunotherapy approaches are under investigation in patients with breast cancer. These include vaccine approaches to elicit strong specific immune responses to tumor antigens such as WT-1, HER2 and NY-ESO-1, approaches involving adoptive transfer of in vitro-expanded, naturally arising or genetically engineered tumor-specific lymphocytes, therapeutic administration of monoclonal antibodies to target and eliminate tumor cells, and approaches that inhibit or destroy the molecular or cellular mediators of cancer-induced immunosuppression, such as CTLA-4, PD-1 or Treg cells. Here we provide a concise and comprehensive review on the role and utility of promising immunotherapeutics for the treatment of patients with breast cancer. </p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"42 ","pages":"30-43"},"PeriodicalIF":0.0,"publicationDate":"2015-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000437183","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"34008635","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
Current Developments in Actively Personalized Cancer Vaccination with a Focus on RNA as the Drug Format. 积极个体化癌症疫苗的最新进展,重点是RNA作为药物格式。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2015-01-01 Epub Date: 2015-09-04 DOI: 10.1159/000437184
Mustafa Diken, Sebastian Kreiter, Björn Kloke, Ugur Sahin

Developments in sequencing technologies have not only led to a rapid generation of genomic and transcriptional data from cancer patients, but also revealed the vast diversity of cancer-specific changes in patient tumors. Among these, mutation changes in the protein sequence can result in novel epitopes recognized by the immune system and, therefore, can be employed in the development of personalized vaccines. Thanks to its easy design and scalable GMP production, vaccines based on mRNAs coding for mutated epitopes have emerged as a reliable strategy for the exploitation of the potential of patient-specific genomic data. In this review, we provide an overview of recent developments in actively personalized vaccinations, with a special focus on the promise of mRNA vaccines.

测序技术的发展不仅导致了癌症患者基因组和转录数据的快速生成,而且揭示了患者肿瘤中癌症特异性变化的巨大多样性。其中,蛋白质序列的突变变化可导致免疫系统识别的新表位,因此可用于个性化疫苗的开发。由于其易于设计和可扩展的GMP生产,基于编码突变表位的mrna的疫苗已成为开发患者特异性基因组数据潜力的可靠策略。在这篇综述中,我们概述了积极个性化疫苗的最新发展,特别关注mRNA疫苗的前景。
{"title":"Current Developments in Actively Personalized Cancer Vaccination with a Focus on RNA as the Drug Format.","authors":"Mustafa Diken,&nbsp;Sebastian Kreiter,&nbsp;Björn Kloke,&nbsp;Ugur Sahin","doi":"10.1159/000437184","DOIUrl":"https://doi.org/10.1159/000437184","url":null,"abstract":"<p><p>Developments in sequencing technologies have not only led to a rapid generation of genomic and transcriptional data from cancer patients, but also revealed the vast diversity of cancer-specific changes in patient tumors. Among these, mutation changes in the protein sequence can result in novel epitopes recognized by the immune system and, therefore, can be employed in the development of personalized vaccines. Thanks to its easy design and scalable GMP production, vaccines based on mRNAs coding for mutated epitopes have emerged as a reliable strategy for the exploitation of the potential of patient-specific genomic data. In this review, we provide an overview of recent developments in actively personalized vaccinations, with a special focus on the promise of mRNA vaccines. </p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"42 ","pages":"44-54"},"PeriodicalIF":0.0,"publicationDate":"2015-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000437184","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"34016100","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 8
Immunotherapies in Early and Advanced Renal Cell Cancer. 早期和晚期肾细胞癌的免疫治疗。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2015-01-01 Epub Date: 2015-09-04 DOI: 10.1159/000436988
Benjamin Kasenda, James Larkin, Martin Gore

The development of new immunomodulatory monoclonal antibodies targeting the CTLA-4 or PD-1 axis has led to a revival of research on immunotherapies in solid tumours including renal cell cancer (RCC). The initial results observed with these monoclonal antibodies in the treatment of advanced melanoma have resulted in considerable interest in this treatment strategy in all tumour types. Preliminary data of these new antibodies in advanced RCC are promising and they have good safety profiles. Response rates are low but durable tumour control has been observed in some patients. However, at the moment there is no evidence that targeting the CTLA-4 or PD-1 axis provides a substantial clinical benefit compared to established treatment with tyrosine kinase or mTOR inhibitors. There are also no reliable predictive markers. At the moment, several randomised trials have been initiated to investigate the new immunomodulatory antibodies either as single agents or in combination with anti-VEGF targeted therapy. Vaccines have continued to be investigated in advanced and adjuvant settings. No trial has so far established vaccines as a standard treatment in either situation. There are still large randomised trials ongoing investigating the potential benefit of a vaccine in combination with standard tyrosine kinase inhibitor therapy. In this chapter we will summarise selected studies on immunotherapy in advanced RCC with a focus on anti-PD-1, anti-PD-L1, and anti-CTLA-4 antibodies. We will also touch briefly on the adjuvant situation and tumour vaccines.

针对CTLA-4或PD-1轴的新型免疫调节单克隆抗体的发展,导致了包括肾细胞癌(RCC)在内的实体肿瘤免疫治疗研究的复兴。这些单克隆抗体治疗晚期黑色素瘤的初步结果引起了人们对这种治疗策略在所有肿瘤类型中的相当大的兴趣。初步数据显示,这些新抗体在晚期肾癌中的应用前景良好,且具有良好的安全性。反应率低,但在一些患者中观察到持久的肿瘤控制。然而,目前没有证据表明,与酪氨酸激酶或mTOR抑制剂的既定治疗相比,靶向CTLA-4或PD-1轴可提供实质性的临床益处。也没有可靠的预测指标。目前,一些随机试验已经启动,以研究新的免疫调节抗体作为单一药物或与抗vegf靶向治疗联合使用。在晚期和佐剂环境中继续研究疫苗。到目前为止,还没有试验将疫苗确定为这两种情况的标准治疗方法。目前仍有大型随机试验正在调查疫苗与标准酪氨酸激酶抑制剂治疗联合使用的潜在益处。在本章中,我们将总结针对晚期RCC的免疫治疗研究,重点是抗pd -1、抗pd - l1和抗ctla -4抗体。我们还将简要介绍佐剂情况和肿瘤疫苗。
{"title":"Immunotherapies in Early and Advanced Renal Cell Cancer.","authors":"Benjamin Kasenda,&nbsp;James Larkin,&nbsp;Martin Gore","doi":"10.1159/000436988","DOIUrl":"https://doi.org/10.1159/000436988","url":null,"abstract":"<p><p>The development of new immunomodulatory monoclonal antibodies targeting the CTLA-4 or PD-1 axis has led to a revival of research on immunotherapies in solid tumours including renal cell cancer (RCC). The initial results observed with these monoclonal antibodies in the treatment of advanced melanoma have resulted in considerable interest in this treatment strategy in all tumour types. Preliminary data of these new antibodies in advanced RCC are promising and they have good safety profiles. Response rates are low but durable tumour control has been observed in some patients. However, at the moment there is no evidence that targeting the CTLA-4 or PD-1 axis provides a substantial clinical benefit compared to established treatment with tyrosine kinase or mTOR inhibitors. There are also no reliable predictive markers. At the moment, several randomised trials have been initiated to investigate the new immunomodulatory antibodies either as single agents or in combination with anti-VEGF targeted therapy. Vaccines have continued to be investigated in advanced and adjuvant settings. No trial has so far established vaccines as a standard treatment in either situation. There are still large randomised trials ongoing investigating the potential benefit of a vaccine in combination with standard tyrosine kinase inhibitor therapy. In this chapter we will summarise selected studies on immunotherapy in advanced RCC with a focus on anti-PD-1, anti-PD-L1, and anti-CTLA-4 antibodies. We will also touch briefly on the adjuvant situation and tumour vaccines. </p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"42 ","pages":"1-10"},"PeriodicalIF":0.0,"publicationDate":"2015-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000436988","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"34010417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 16
Successes and limitations of targeted cancer therapy in colon cancer. 结肠癌靶向治疗的成功与局限性。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2014-01-01 Epub Date: 2014-02-17 DOI: 10.1159/000356436
Claus-Henning Köhne

Constant development of chemotherapy and more recently the introduction of VEGF- and epidermal growth factor receptor (EGFR)-directed agents have improved significantly the treatment of patients with colorectal cancer. In the adjuvant setting, especially for UICC stage III colon cancer patients, fluoropyrimidine in combination with oxaliplatin is usually the standard of care. With some surprise, both VEGF inhibitors (for all patients) and EGFR (for patients with KRAS exon 2 mutant tumors) have failed to improve adjuvant chemotherapy. Also, adding an EGFR antibody to FOLFOX as perioperative treatment in patients with resectable exon 2 KRAS wild-type liver metastases was not successful. However, patients with metastatic disease harboring a RAS wild-type tumor are with no doubt candidates for combination chemotherapy plus an EGFR antibody. In patients with liver-limited disease, metastases may become resectable following intensive chemotherapy (including an EGFR antibody for RAS wild-type disease), which may result in cure or significantly prolonged survival. In the case of RAS wild-type tumors, median survival in patients with unresectable metastases approaches now 3 years if EGFR antibodies are used in the first line. There is little evidence for VEGF inhibitors in patients with RAS wild-type or mutant disease in first-line chemotherapy if combination chemotherapy is considered. VEGF inhibitors, however, are very potent drugs to be combined with chemotherapy for second-line treatment.

化疗的不断发展以及近年来VEGF和表皮生长因子受体(EGFR)导向药物的引入显著改善了结直肠癌患者的治疗。在辅助治疗中,特别是对于UICC III期结肠癌患者,氟嘧啶联合奥沙利铂通常是标准的治疗方案。令人惊讶的是,VEGF抑制剂(适用于所有患者)和EGFR(适用于KRAS外显子2突变肿瘤患者)都未能改善辅助化疗。此外,将EGFR抗体加入FOLFOX作为可切除外显子2 KRAS野生型肝转移患者的围手术期治疗并不成功。然而,携带RAS野生型肿瘤的转移性疾病患者无疑是联合化疗加EGFR抗体的候选者。在肝局限性疾病患者中,在强化化疗(包括针对RAS野生型疾病的EGFR抗体)后,转移灶可以切除,这可能导致治愈或显著延长生存期。在RAS野生型肿瘤的情况下,如果在一线使用EGFR抗体,不可切除转移的患者的中位生存期现在接近3年。如果考虑联合化疗,在一线化疗中RAS野生型或突变型疾病患者中VEGF抑制剂的作用几乎没有证据。然而,VEGF抑制剂是一种非常有效的药物,可以与化疗联合用于二线治疗。
{"title":"Successes and limitations of targeted cancer therapy in colon cancer.","authors":"Claus-Henning Köhne","doi":"10.1159/000356436","DOIUrl":"https://doi.org/10.1159/000356436","url":null,"abstract":"<p><p>Constant development of chemotherapy and more recently the introduction of VEGF- and epidermal growth factor receptor (EGFR)-directed agents have improved significantly the treatment of patients with colorectal cancer. In the adjuvant setting, especially for UICC stage III colon cancer patients, fluoropyrimidine in combination with oxaliplatin is usually the standard of care. With some surprise, both VEGF inhibitors (for all patients) and EGFR (for patients with KRAS exon 2 mutant tumors) have failed to improve adjuvant chemotherapy. Also, adding an EGFR antibody to FOLFOX as perioperative treatment in patients with resectable exon 2 KRAS wild-type liver metastases was not successful. However, patients with metastatic disease harboring a RAS wild-type tumor are with no doubt candidates for combination chemotherapy plus an EGFR antibody. In patients with liver-limited disease, metastases may become resectable following intensive chemotherapy (including an EGFR antibody for RAS wild-type disease), which may result in cure or significantly prolonged survival. In the case of RAS wild-type tumors, median survival in patients with unresectable metastases approaches now 3 years if EGFR antibodies are used in the first line. There is little evidence for VEGF inhibitors in patients with RAS wild-type or mutant disease in first-line chemotherapy if combination chemotherapy is considered. VEGF inhibitors, however, are very potent drugs to be combined with chemotherapy for second-line treatment.</p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"41 ","pages":"36-50"},"PeriodicalIF":0.0,"publicationDate":"2014-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000356436","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"32259452","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 21
Successes and limitations of targeted cancer therapy in lung cancer. 肺癌靶向治疗的成功与局限。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2014-01-01 Epub Date: 2014-02-17 DOI: 10.1159/000355902
Kenichi Suda, Tetsuya Mitsudomi

Human cancers usually evolve through multistep processes. These processes are driven by the accumulation of abundant genetic and epigenetic abnormalities. However, some lung cancers depend on a single activated oncogene by somatic mutation, termed 'driver oncogenic mutations', for their proliferation and survival. EGFR(epidermal growth factor receptor) mutations and ALK(anaplastic lymphoma kinase) rearrangement are typical examples of such driver oncogenic mutations found in lung adenocarcinomas. EGFR-tyrosine kinase inhibitors (TKIs) or ALK-TKIs significantly improved treatment outcomes compared with conventional cytotoxic chemotherapy in patients with lung cancers harboring EGFR mutations or ALK rearrangement, respectively. Therefore, treatment strategies for lung cancers have dramatically changed from a 'general and empiric' to a 'personalized and evidence-based' approach according to the driver oncogenic mutation. Several novel driver oncogenic mutations, which are candidates as novel targets, such as ERBB2, BRAF, ROS1, and RET, have been discovered. Despite these successes, several limitations have arisen. One example is that some lung cancers do not respond to treatments targeting driver oncogenic mutations, as exemplified in KRAS-mutated lung cancers. Another is resistance to molecular-targeted drugs. Such resistance includes de novo resistance and acquired resistance. A number of molecular mechanisms underlying such resistance have been reported. These mechanisms can be roughly divided into three categories: alteration of the targeted oncogenes themselves by secondary mutations or amplification, activation of an alternative oncogenic signaling track, and conversion of cellular characteristics. Overcoming resistance is a current area of urgent clinical research.

人类癌症通常是通过多个步骤进化而来的。这些过程是由大量遗传和表观遗传异常的积累所驱动的。然而,一些肺癌依赖于通过体细胞突变激活的单个致癌基因,称为“驱动致癌突变”,以促进其增殖和存活。EGFR(表皮生长因子受体)突变和ALK(间变性淋巴瘤激酶)重排是在肺腺癌中发现的这种驱动致癌突变的典型例子。与传统细胞毒性化疗相比,EGFR-酪氨酸激酶抑制剂(TKIs)或ALK-TKIs分别显著改善了EGFR突变或ALK重排肺癌患者的治疗结果。因此,肺癌的治疗策略已经发生了巨大的变化,从“一般和经验”到“个性化和基于证据”的方法,根据驱动致癌突变。已经发现了一些新的驱动致癌突变,如ERBB2、BRAF、ROS1和RET,它们是新靶点的候选者。尽管取得了这些成功,但也出现了一些限制。一个例子是,一些肺癌对针对驱动致癌突变的治疗没有反应,如kras突变的肺癌。另一个是对分子靶向药物的耐药性。这种抗性包括新生抗性和获得性抗性。已经报道了这种耐药性背后的一些分子机制。这些机制大致可分为三类:通过继发性突变或扩增改变靶癌基因本身,激活另一种致癌信号通路,以及细胞特性的转化。克服耐药性是当前迫切需要临床研究的领域。
{"title":"Successes and limitations of targeted cancer therapy in lung cancer.","authors":"Kenichi Suda,&nbsp;Tetsuya Mitsudomi","doi":"10.1159/000355902","DOIUrl":"https://doi.org/10.1159/000355902","url":null,"abstract":"<p><p>Human cancers usually evolve through multistep processes. These processes are driven by the accumulation of abundant genetic and epigenetic abnormalities. However, some lung cancers depend on a single activated oncogene by somatic mutation, termed 'driver oncogenic mutations', for their proliferation and survival. EGFR(epidermal growth factor receptor) mutations and ALK(anaplastic lymphoma kinase) rearrangement are typical examples of such driver oncogenic mutations found in lung adenocarcinomas. EGFR-tyrosine kinase inhibitors (TKIs) or ALK-TKIs significantly improved treatment outcomes compared with conventional cytotoxic chemotherapy in patients with lung cancers harboring EGFR mutations or ALK rearrangement, respectively. Therefore, treatment strategies for lung cancers have dramatically changed from a 'general and empiric' to a 'personalized and evidence-based' approach according to the driver oncogenic mutation. Several novel driver oncogenic mutations, which are candidates as novel targets, such as ERBB2, BRAF, ROS1, and RET, have been discovered. Despite these successes, several limitations have arisen. One example is that some lung cancers do not respond to treatments targeting driver oncogenic mutations, as exemplified in KRAS-mutated lung cancers. Another is resistance to molecular-targeted drugs. Such resistance includes de novo resistance and acquired resistance. A number of molecular mechanisms underlying such resistance have been reported. These mechanisms can be roughly divided into three categories: alteration of the targeted oncogenes themselves by secondary mutations or amplification, activation of an alternative oncogenic signaling track, and conversion of cellular characteristics. Overcoming resistance is a current area of urgent clinical research.</p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"41 ","pages":"62-77"},"PeriodicalIF":0.0,"publicationDate":"2014-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000355902","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"32259454","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 37
Successes and limitations of targeted therapies in renal cell carcinoma. 肾细胞癌靶向治疗的成功与局限性。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2014-01-01 Epub Date: 2014-02-17 DOI: 10.1159/000355906
Marc Pracht, Dominik Berthold

Until recently, the standard treatment for metastatic renal cell carcinoma (RCC) was nonspecific immunotherapy based on interleukin-2 or interferon-α. This was associated with a modest survival benefit and with significant clinical toxicities. The understanding of numerous molecular pathways in RCC, including HIF, VEGF, mTOR, and the consecutive use of targeted therapies since the beginning of 2005 have significantly improved outcomes for patients with metastatic RCC with an overall survival greater than 2 years. At present, at least 7 targeted agents are approved for first and consecutive lines of treatment of clear cell metastatic RCC. Long-term benefit and extended survival may be achieved through the optimal use of targeted therapies: optimal dosing, adverse event management and treatment duration and compliance. Advances in the finding of prognostic factors highlight the potential for personalizing treatment for patients with metastatic RCC. Data regarding the best sequencing of targeted therapies, predictive biomarkers, best timing of surgery, patient risk profiles, understanding of resistance mechanisms and safety of targeted therapies are growing and will provide a further step ahead in the management of advanced RCC. In parallel, a new class of therapeutics is emerging in RCC: immunotherapy; in particular check-point blockade antibodies are showing very promising results.

直到最近,转移性肾细胞癌(RCC)的标准治疗是基于白细胞介素-2或干扰素-α的非特异性免疫治疗。这与适度的生存获益和显著的临床毒性相关。对包括HIF、VEGF、mTOR在内的许多RCC分子通路的了解,以及自2005年初以来连续使用靶向治疗,显著改善了总生存期大于2年的转移性RCC患者的预后。目前,至少有7种靶向药物被批准用于透明细胞转移性RCC的一线和连续治疗。通过最佳使用靶向治疗:最佳剂量、不良事件管理、治疗持续时间和依从性,可以实现长期获益和延长生存期。在发现预后因素方面的进展突出了转移性肾细胞癌患者个性化治疗的潜力。关于靶向治疗的最佳测序、预测性生物标志物、最佳手术时机、患者风险概况、对耐药机制的理解和靶向治疗的安全性的数据正在增长,并将为晚期RCC的管理提供进一步的进展。同时,一种新的治疗方法正在RCC中出现:免疫治疗;特别是检查点阻断抗体显示出非常有希望的结果。
{"title":"Successes and limitations of targeted therapies in renal cell carcinoma.","authors":"Marc Pracht,&nbsp;Dominik Berthold","doi":"10.1159/000355906","DOIUrl":"https://doi.org/10.1159/000355906","url":null,"abstract":"<p><p>Until recently, the standard treatment for metastatic renal cell carcinoma (RCC) was nonspecific immunotherapy based on interleukin-2 or interferon-α. This was associated with a modest survival benefit and with significant clinical toxicities. The understanding of numerous molecular pathways in RCC, including HIF, VEGF, mTOR, and the consecutive use of targeted therapies since the beginning of 2005 have significantly improved outcomes for patients with metastatic RCC with an overall survival greater than 2 years. At present, at least 7 targeted agents are approved for first and consecutive lines of treatment of clear cell metastatic RCC. Long-term benefit and extended survival may be achieved through the optimal use of targeted therapies: optimal dosing, adverse event management and treatment duration and compliance. Advances in the finding of prognostic factors highlight the potential for personalizing treatment for patients with metastatic RCC. Data regarding the best sequencing of targeted therapies, predictive biomarkers, best timing of surgery, patient risk profiles, understanding of resistance mechanisms and safety of targeted therapies are growing and will provide a further step ahead in the management of advanced RCC. In parallel, a new class of therapeutics is emerging in RCC: immunotherapy; in particular check-point blockade antibodies are showing very promising results.</p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"41 ","pages":"98-112"},"PeriodicalIF":0.0,"publicationDate":"2014-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000355906","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"32259457","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 13
Successes and limitations of targeted cancer therapy in gastrointestinal stromal tumors. 靶向肿瘤治疗胃肠道间质瘤的成功与局限性。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2014-01-01 Epub Date: 2014-02-17 DOI: 10.1159/000355898
Paolo G Casali

In gastrointestinal stromal tumors (GIST), molecularly targeted therapies, starting with imatinib, are directed against an activating mutation of KIT or PDGFRA, which drives the disease. Their efficacy has brought the median survival from one to at least 5 years in the metastatic setting. Tumor response patterns may include tumor shrinkage or not, but are marked by pathologic and radiological changes in tumor tissue. Tumor sensitivity to imatinib can be precisely predicted by the mutational status. However, the metastatic disease has not been truly converted into a chronic condition since secondary resistance to imatinib remains a major limiting factor occurring after a median of 2 years at least in most patients. Further-line therapies are available, i.e. with sunitinib and regorafenib, which can prolong progression-free survival for limited time intervals. Resistance is due to secondary mutations. These give rise to a molecular heterogeneity, which represents a formidable therapeutic challenge. However, the scenario has aspects of a 'liquid resistance'. In fact, resistance may spread in a stepwise fashion throughout the tumor: focal progression may be one possible clinical presentation, and tyrosine kinase inhibitors may impact tumor growth even beyond conventional progression. In addition, sensitive and resistant clones may expand and shrink depending on the selective pressure of tyrosine kinase inhibitors, with be possible responses on rechallenge with drugs. In the adjuvant treatment of high-risk molecularly sensitive GIST, imatinib is able to substantially delay relapses, if due to occur, with a limited survival benefit, though, apparently, without impacting the cure rate.

在胃肠道间质瘤(GIST)中,从伊马替尼开始的分子靶向治疗针对KIT或PDGFRA的激活突变,这是导致疾病的原因。它们的疗效使转移性肿瘤的中位生存期从1年延长到至少5年。肿瘤反应模式可能包括肿瘤缩小或不缩小,但以肿瘤组织的病理和放射学变化为标志。肿瘤对伊马替尼的敏感性可以通过突变状态精确预测。然而,转移性疾病并没有真正转化为慢性疾病,因为对伊马替尼的继发性耐药仍然是主要的限制因素,至少在大多数患者中位2年后发生。进一步的一线治疗是可用的,即舒尼替尼和瑞非尼,可以延长有限时间间隔的无进展生存期。抗性是由于继发性突变。这些导致了分子异质性,这是一个巨大的治疗挑战。然而,这种情况有“液体阻力”的方面。事实上,耐药可能在整个肿瘤中逐步扩散:局灶性进展可能是一种可能的临床表现,酪氨酸激酶抑制剂可能影响肿瘤生长,甚至超出常规进展。此外,敏感和耐药克隆可能会根据酪氨酸激酶抑制剂的选择压力而扩大和缩小,并可能对药物的再挑战产生反应。在高危分子敏感GIST的辅助治疗中,伊马替尼能够显著延缓复发(如果发生的话),虽然明显不影响治愈率,但生存获益有限。
{"title":"Successes and limitations of targeted cancer therapy in gastrointestinal stromal tumors.","authors":"Paolo G Casali","doi":"10.1159/000355898","DOIUrl":"https://doi.org/10.1159/000355898","url":null,"abstract":"<p><p>In gastrointestinal stromal tumors (GIST), molecularly targeted therapies, starting with imatinib, are directed against an activating mutation of KIT or PDGFRA, which drives the disease. Their efficacy has brought the median survival from one to at least 5 years in the metastatic setting. Tumor response patterns may include tumor shrinkage or not, but are marked by pathologic and radiological changes in tumor tissue. Tumor sensitivity to imatinib can be precisely predicted by the mutational status. However, the metastatic disease has not been truly converted into a chronic condition since secondary resistance to imatinib remains a major limiting factor occurring after a median of 2 years at least in most patients. Further-line therapies are available, i.e. with sunitinib and regorafenib, which can prolong progression-free survival for limited time intervals. Resistance is due to secondary mutations. These give rise to a molecular heterogeneity, which represents a formidable therapeutic challenge. However, the scenario has aspects of a 'liquid resistance'. In fact, resistance may spread in a stepwise fashion throughout the tumor: focal progression may be one possible clinical presentation, and tyrosine kinase inhibitors may impact tumor growth even beyond conventional progression. In addition, sensitive and resistant clones may expand and shrink depending on the selective pressure of tyrosine kinase inhibitors, with be possible responses on rechallenge with drugs. In the adjuvant treatment of high-risk molecularly sensitive GIST, imatinib is able to substantially delay relapses, if due to occur, with a limited survival benefit, though, apparently, without impacting the cure rate.</p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"41 ","pages":"51-61"},"PeriodicalIF":0.0,"publicationDate":"2014-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000355898","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"32259453","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 14
Successes and limitations of targeted cancer therapy in melanoma. 靶向治疗黑色素瘤的成功与局限。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2014-01-01 Epub Date: 2014-02-17 DOI: 10.1159/000355903
Emanuela Romano, Olivier Michielin

The treatment of stage IV melanoma has witnessed a very impressive pace of innovation in recent years, to a point where the management of these patients has very little in common to what was standard practice 5 years ago. If the gain in overall survival, the high response rates or the induction of a significant fraction of long survivors are all very exciting news for our patients and their families, the path that led to these discoveries is as important. Rather than empirical, the development of these new strategies has been extremely rational, based on state-of-the-art basic biology and immunology, exemplary translational research and, finally, hypothesis-driven targeted trials that led to rapid approval. In this review, we will cover all the new targeted therapies that have emerged as the results of these translational programs, focusing mainly on signaling pathway- and immune checkpoint-targeted therapies. Taken collectively, these new developments set the bar for a new paradigm in future translational and clinical research in both melanoma as well as other tumor types.

近年来,IV期黑色素瘤的治疗经历了令人印象深刻的创新步伐,以至于这些患者的管理与5年前的标准实践几乎没有什么共同之处。如果总体生存率的提高,高反应率或诱导相当一部分长期幸存者对我们的患者和他们的家人来说都是非常令人兴奋的消息,那么导致这些发现的途径同样重要。这些新策略的发展非常理性,基于最先进的基础生物学和免疫学,典型的转化研究,以及最终导致快速批准的假设驱动的靶向试验,而不是经验。在这篇综述中,我们将涵盖作为这些转化项目的结果而出现的所有新的靶向治疗,主要关注信号通路和免疫检查点靶向治疗。总的来说,这些新的发展为黑色素瘤和其他肿瘤类型的未来转化和临床研究树立了新的典范。
{"title":"Successes and limitations of targeted cancer therapy in melanoma.","authors":"Emanuela Romano,&nbsp;Olivier Michielin","doi":"10.1159/000355903","DOIUrl":"https://doi.org/10.1159/000355903","url":null,"abstract":"<p><p>The treatment of stage IV melanoma has witnessed a very impressive pace of innovation in recent years, to a point where the management of these patients has very little in common to what was standard practice 5 years ago. If the gain in overall survival, the high response rates or the induction of a significant fraction of long survivors are all very exciting news for our patients and their families, the path that led to these discoveries is as important. Rather than empirical, the development of these new strategies has been extremely rational, based on state-of-the-art basic biology and immunology, exemplary translational research and, finally, hypothesis-driven targeted trials that led to rapid approval. In this review, we will cover all the new targeted therapies that have emerged as the results of these translational programs, focusing mainly on signaling pathway- and immune checkpoint-targeted therapies. Taken collectively, these new developments set the bar for a new paradigm in future translational and clinical research in both melanoma as well as other tumor types.</p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"41 ","pages":"78-88"},"PeriodicalIF":0.0,"publicationDate":"2014-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000355903","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"32259455","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Successes and limitations of targeted cancer therapy in breast cancer. 乳腺癌靶向治疗的成功与局限性。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2014-01-01 Epub Date: 2014-02-17 DOI: 10.1159/000355896
Giuseppe Curigliano, Carmen Criscitiello

Breast cancer is not a single disease. Specific biological processes and distinct genetic pathways are associated with prognosis and sensitivity to chemotherapy and targeted agents in different subtypes of breast cancers. As a consequence, breast cancer can be classified by molecular events. A primary challenge for future drug development in breast cancer will be to distinguish genes and pathways that 'drive' cancer proliferation (drivers) from genes and pathways that have no role in the development of cancer (passengers). The identification of functional pathways that are enriched for mutated genes will select subpopulation of patients likely to be sensitive to biology-driven targeted agents. The selection of driver pathways in resistant tumors will permit to discover a biology-driven platform for new drug development to overcome resistance. We are moving in the era of stratified and personalized therapy. Personalized cancer therapy is based on the precept that detailed molecular characterization of the patient's tumor and its microenvironment will enable tailored therapies to improve outcomes and decrease toxicity. However, there are numerous challenges we need to overcome before delivering on the promise of personalized cancer therapy. These include tumor heterogeneity and molecular evolution, costs and potential morbidity of biopsies, lack of effective drugs against most genomic aberrations, technical limitations of molecular tests, and reimbursement and regulatory hurdles. Critically, successes and limitations surrounding personalized cancer therapy must be tempered with realistic expectations, which, today, encompass increased survival times for only a portion of patients.

乳腺癌不是一种单一的疾病。特定的生物学过程和独特的遗传途径与不同亚型乳腺癌的预后和对化疗和靶向药物的敏感性有关。因此,乳腺癌可以根据分子事件进行分类。未来乳腺癌药物开发的一个主要挑战将是区分“驱动”癌症增殖的基因和途径(驱动因素)与在癌症发展中没有作用的基因和途径(乘客因素)。鉴定富含突变基因的功能通路将选择可能对生物学驱动的靶向药物敏感的患者亚群。耐药肿瘤中驱动途径的选择将允许发现一个生物学驱动的新药开发平台,以克服耐药性。我们正在进入分层和个性化治疗的时代。个性化癌症治疗是基于这样的原则:对患者肿瘤及其微环境进行详细的分子表征,将使量身定制的治疗能够改善结果并降低毒性。然而,在实现个性化癌症治疗的承诺之前,我们需要克服许多挑战。这些挑战包括肿瘤异质性和分子进化、活组织检查的成本和潜在发病率、缺乏针对大多数基因组畸变的有效药物、分子检测的技术限制以及报销和监管障碍。关键的是,个性化癌症治疗的成功和局限性必须与现实的期望相调和,今天,只有一部分患者的生存时间增加。
{"title":"Successes and limitations of targeted cancer therapy in breast cancer.","authors":"Giuseppe Curigliano,&nbsp;Carmen Criscitiello","doi":"10.1159/000355896","DOIUrl":"https://doi.org/10.1159/000355896","url":null,"abstract":"<p><p>Breast cancer is not a single disease. Specific biological processes and distinct genetic pathways are associated with prognosis and sensitivity to chemotherapy and targeted agents in different subtypes of breast cancers. As a consequence, breast cancer can be classified by molecular events. A primary challenge for future drug development in breast cancer will be to distinguish genes and pathways that 'drive' cancer proliferation (drivers) from genes and pathways that have no role in the development of cancer (passengers). The identification of functional pathways that are enriched for mutated genes will select subpopulation of patients likely to be sensitive to biology-driven targeted agents. The selection of driver pathways in resistant tumors will permit to discover a biology-driven platform for new drug development to overcome resistance. We are moving in the era of stratified and personalized therapy. Personalized cancer therapy is based on the precept that detailed molecular characterization of the patient's tumor and its microenvironment will enable tailored therapies to improve outcomes and decrease toxicity. However, there are numerous challenges we need to overcome before delivering on the promise of personalized cancer therapy. These include tumor heterogeneity and molecular evolution, costs and potential morbidity of biopsies, lack of effective drugs against most genomic aberrations, technical limitations of molecular tests, and reimbursement and regulatory hurdles. Critically, successes and limitations surrounding personalized cancer therapy must be tempered with realistic expectations, which, today, encompass increased survival times for only a portion of patients.</p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"41 ","pages":"15-35"},"PeriodicalIF":0.0,"publicationDate":"2014-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000355896","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"32258502","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 30
Successes and limitations of targeted cancer therapy in ovarian cancer. 卵巢癌靶向治疗的成功与局限性。
Q4 Biochemistry, Genetics and Molecular Biology Pub Date : 2014-01-01 Epub Date: 2014-02-17 DOI: 10.1159/000355905
Giovanna Damia, Cristiana Sessa

In ovarian cancer, the clinical development of anticancer agents targeting DNA repair has been associated with significant results because of the elucidation of the different types of damages and repair systems, including PARP. The discovery of the BRCA mutation and its role in ovarian cancer and the clinical application of the concept of synthetic lethality have been the rationale for the successful testing of PARP inhibitors in BRCA mutated ovarian cancer patients. The recent knowledge of the molecular features of low grade ovarian cancer and the application of the concept of synthetic lethality also in this well-defined pathological entity have prompted the clinical evaluation of a combination of PI3K/MEK inhibitors, the first results of which have been already reported.

在卵巢癌中,靶向DNA修复的抗癌药物的临床开发已经取得了显著的成果,因为阐明了不同类型的损伤和修复系统,包括PARP。BRCA突变及其在卵巢癌中的作用的发现以及合成致死概念的临床应用是PARP抑制剂在BRCA突变卵巢癌患者中成功测试的基本原理。最近对低级别卵巢癌分子特征的了解,以及在这种明确定义的病理实体中应用合成致死性概念,促使对PI3K/MEK抑制剂组合的临床评估,其第一个结果已经报道。
{"title":"Successes and limitations of targeted cancer therapy in ovarian cancer.","authors":"Giovanna Damia,&nbsp;Cristiana Sessa","doi":"10.1159/000355905","DOIUrl":"https://doi.org/10.1159/000355905","url":null,"abstract":"<p><p>In ovarian cancer, the clinical development of anticancer agents targeting DNA repair has been associated with significant results because of the elucidation of the different types of damages and repair systems, including PARP. The discovery of the BRCA mutation and its role in ovarian cancer and the clinical application of the concept of synthetic lethality have been the rationale for the successful testing of PARP inhibitors in BRCA mutated ovarian cancer patients. The recent knowledge of the molecular features of low grade ovarian cancer and the application of the concept of synthetic lethality also in this well-defined pathological entity have prompted the clinical evaluation of a combination of PI3K/MEK inhibitors, the first results of which have been already reported.</p>","PeriodicalId":49661,"journal":{"name":"Progress in Tumor Research","volume":"41 ","pages":"89-97"},"PeriodicalIF":0.0,"publicationDate":"2014-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1159/000355905","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"32259456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 9
期刊
Progress in Tumor Research
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