首页 > 最新文献

Expert Review of Precision Medicine and Drug Development最新文献

英文 中文
Genomic sequencing in severe epilepsy: a step closer to precision medicine 严重癫痫的基因组测序:向精准医学迈进了一步
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-02-25 DOI: 10.1080/23808993.2020.1732203
M. Esposito, Ilaria Lagorio, D. Peroni, A. Bonuccelli, A. Orsini, P. Striano
ABSTRACT Introduction: The large number of different syndromes and seizure types, together with an inter-individual variable response to antiepileptic drugs (AEDs), makes the treatment of epilepsy challenging. Areas covered: Early infantile Epileptic encephalopathies (EIEE) are a group of neurodevelopmental disorders consisting of early-onset refractory seizures often accompanied by important developmental delay or regression. The last two decades have seen major advancement in the diagnosis of epilepsy thanks both to modern neuroimaging but, primarily, to new methods in molecular genetics and gene sequencing. The application of Next-Generation Sequencing (NGS) techniques has already helped to understand the genetic diversity and underlying pathogenic mechanisms in severe epilepsy of childhood. Furthermore, the use of Whole Exome Sequencing (WES) in parent–offspring trios has also helped to identify de novo mutations in patients with EIEE. Tailored treatments are already applicable in some monogenic epilepsy, but these are a minority of cases. Expert commentary: In our opinion, the future of epilepsy treatment will be multidisciplinary and possibly very different from the currently almost empiric approach and we are confident that a ‘precision medicine’ will be applicable on large scale.
摘要:大量不同的综合征和癫痫发作类型,以及个体间对抗癫痫药物(aed)的不同反应,使得癫痫的治疗具有挑战性。涵盖领域:早期婴儿癫痫性脑病(eee)是一组神经发育障碍,包括早发性难治性癫痫发作,通常伴有重要的发育迟缓或倒退。在过去的二十年中,癫痫的诊断取得了重大进展,这不仅归功于现代神经成像,而且主要归功于分子遗传学和基因测序的新方法。新一代测序(NGS)技术的应用已经有助于了解儿童严重癫痫的遗传多样性和潜在致病机制。此外,使用全外显子组测序(WES)在父母-后代三人组中也有助于识别eee患者的新生突变。量身定制的治疗方法已经适用于一些单基因癫痫,但这只是少数病例。专家评论:在我们看来,未来的癫痫治疗将是多学科的,可能与目前几乎是经验主义的方法大不相同,我们有信心“精准医学”将大规模适用。
{"title":"Genomic sequencing in severe epilepsy: a step closer to precision medicine","authors":"M. Esposito, Ilaria Lagorio, D. Peroni, A. Bonuccelli, A. Orsini, P. Striano","doi":"10.1080/23808993.2020.1732203","DOIUrl":"https://doi.org/10.1080/23808993.2020.1732203","url":null,"abstract":"ABSTRACT Introduction: The large number of different syndromes and seizure types, together with an inter-individual variable response to antiepileptic drugs (AEDs), makes the treatment of epilepsy challenging. Areas covered: Early infantile Epileptic encephalopathies (EIEE) are a group of neurodevelopmental disorders consisting of early-onset refractory seizures often accompanied by important developmental delay or regression. The last two decades have seen major advancement in the diagnosis of epilepsy thanks both to modern neuroimaging but, primarily, to new methods in molecular genetics and gene sequencing. The application of Next-Generation Sequencing (NGS) techniques has already helped to understand the genetic diversity and underlying pathogenic mechanisms in severe epilepsy of childhood. Furthermore, the use of Whole Exome Sequencing (WES) in parent–offspring trios has also helped to identify de novo mutations in patients with EIEE. Tailored treatments are already applicable in some monogenic epilepsy, but these are a minority of cases. Expert commentary: In our opinion, the future of epilepsy treatment will be multidisciplinary and possibly very different from the currently almost empiric approach and we are confident that a ‘precision medicine’ will be applicable on large scale.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 1","pages":"101 - 108"},"PeriodicalIF":1.2,"publicationDate":"2020-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1732203","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43865201","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: 4
Renal Cell Carcinoma: genomic landscape and clinical implications 肾细胞癌:基因组景观和临床意义
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-02-24 DOI: 10.1080/23808993.2020.1733407
G. Aurilio, M. Santoni, A. Cimadamore, F. Massari, M. Scarpelli, A. López-Beltran, Liang Cheng, N. Battelli, F. Nolè, R. Montironi
ABSTRACT Introduction: The route to precision medicine in Renal Cell Carcinoma (RCC) is still full of challenges for worldwide uro-oncologists. This is mainly related to the high complexity of the genomic landscape of this tumor. Area covered: In this review, we focused on the most recent advances on RCC genomic scenario and its clinical and prognostic implications. In particular, we describe the main gene alterations that occur during RCC development and progression. At this purpose, we extensively analyzed the available literature from Pubmed archives on this field. Expert commentary: Summarizing all available data and taking separately each putative biomarker illustrated, we feel to conclude that there is a certain correlation between the alterations occurring in BAP1, PBMR1, and SETD2 genes and the prognosis of RCC patients. However, apart from this individual analysis, the mutational scenario in RCC seems to be even more intricate. The evolution of RCC will pass through the optimization of emerging laboratory techniques and to a progressive integration of these methodologies within daily clinical practice and in the context of randomized trials.
导读:肾细胞癌(RCC)的精准医疗之路对全球泌尿肿瘤学家来说仍然充满挑战。这主要与这种肿瘤的基因组景观的高度复杂性有关。涵盖领域:在这篇综述中,我们重点介绍了RCC基因组情景及其临床和预后意义的最新进展。特别是,我们描述的主要基因改变,发生在RCC的发展和进展。为此,我们广泛分析了Pubmed档案中关于这一领域的现有文献。专家评论:总结所有可用的数据,并单独考虑每个假定的生物标志物,我们认为BAP1, PBMR1和SETD2基因的改变与RCC患者的预后之间存在一定的相关性。然而,除了这种个体分析之外,RCC的突变情况似乎更加复杂。RCC的发展将通过新兴实验室技术的优化以及这些方法在日常临床实践和随机试验背景下的逐步整合。
{"title":"Renal Cell Carcinoma: genomic landscape and clinical implications","authors":"G. Aurilio, M. Santoni, A. Cimadamore, F. Massari, M. Scarpelli, A. López-Beltran, Liang Cheng, N. Battelli, F. Nolè, R. Montironi","doi":"10.1080/23808993.2020.1733407","DOIUrl":"https://doi.org/10.1080/23808993.2020.1733407","url":null,"abstract":"ABSTRACT Introduction: The route to precision medicine in Renal Cell Carcinoma (RCC) is still full of challenges for worldwide uro-oncologists. This is mainly related to the high complexity of the genomic landscape of this tumor. Area covered: In this review, we focused on the most recent advances on RCC genomic scenario and its clinical and prognostic implications. In particular, we describe the main gene alterations that occur during RCC development and progression. At this purpose, we extensively analyzed the available literature from Pubmed archives on this field. Expert commentary: Summarizing all available data and taking separately each putative biomarker illustrated, we feel to conclude that there is a certain correlation between the alterations occurring in BAP1, PBMR1, and SETD2 genes and the prognosis of RCC patients. However, apart from this individual analysis, the mutational scenario in RCC seems to be even more intricate. The evolution of RCC will pass through the optimization of emerging laboratory techniques and to a progressive integration of these methodologies within daily clinical practice and in the context of randomized trials.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 1","pages":"100 - 95"},"PeriodicalIF":1.2,"publicationDate":"2020-02-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1733407","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48532608","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: 1
The emerging role of precision medicine in the treatment of endometrial cancer 精准医疗在子宫内膜癌症治疗中的新作用
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-02-20 DOI: 10.1080/23808993.2020.1732204
M. Toboni, D. Mutch
ABSTRACT Introduction: Uterine cancer is the most common gynecologic malignancy with a high cure rate for early stage disease. Surgical and standard chemotherapy have provided modest results. Precision therapies targeting molecular pathways have recently produced encouraging results. Areas covered: This paper describes molecular pathways including PI3 K/AKT/mTor, VEGF, and angiogenesis, HER2/neu and EGFR, various endocrine therapies (including megace, medroxyprogesterone acetate, tamoxifen, letrozole, and anastrozole) and immune checkpoint inhibitors. An extensive literature search was completed including but not limited to PubMed, Google Scholar, and the studies conducted by the Gynecologic Oncology Group (GOG), to identify all relevant clinical trials opened to assess clinical benefit in patients with advanced endometrial cancer. Expert commentary: Molecular characterization of tumors provides an opportunity to tailor therapy to individual patients. With continual innovation, eventually real-time changes in treatment regimens can be made for individual patients to treat the ever-changing molecular landscape of the tumor. Immunotherapy remains the most promising treatment for advanced endometrial cancers.
摘要简介:子宫癌是妇科最常见的恶性肿瘤,早期治愈率高。手术和标准化疗的效果一般。针对分子途径的精确治疗最近产生了令人鼓舞的结果。涉及领域:本文描述了分子通路,包括pi3k /AKT/mTor, VEGF和血管生成,HER2/neu和EGFR,各种内分泌疗法(包括megace,醋酸甲孕酮,他莫昔芬,来曲唑和阿那曲唑)和免疫检查点抑制剂。我们完成了广泛的文献检索,包括但不限于PubMed、b谷歌Scholar和妇科肿瘤组(GOG)进行的研究,以确定所有相关的临床试验,以评估晚期子宫内膜癌患者的临床获益。专家评论:肿瘤的分子特征提供了一个机会,量身定制治疗的个别患者。随着不断的创新,最终可以为个体患者实时改变治疗方案,以治疗不断变化的肿瘤分子景观。免疫疗法仍然是晚期子宫内膜癌最有希望的治疗方法。
{"title":"The emerging role of precision medicine in the treatment of endometrial cancer","authors":"M. Toboni, D. Mutch","doi":"10.1080/23808993.2020.1732204","DOIUrl":"https://doi.org/10.1080/23808993.2020.1732204","url":null,"abstract":"ABSTRACT Introduction: Uterine cancer is the most common gynecologic malignancy with a high cure rate for early stage disease. Surgical and standard chemotherapy have provided modest results. Precision therapies targeting molecular pathways have recently produced encouraging results. Areas covered: This paper describes molecular pathways including PI3 K/AKT/mTor, VEGF, and angiogenesis, HER2/neu and EGFR, various endocrine therapies (including megace, medroxyprogesterone acetate, tamoxifen, letrozole, and anastrozole) and immune checkpoint inhibitors. An extensive literature search was completed including but not limited to PubMed, Google Scholar, and the studies conducted by the Gynecologic Oncology Group (GOG), to identify all relevant clinical trials opened to assess clinical benefit in patients with advanced endometrial cancer. Expert commentary: Molecular characterization of tumors provides an opportunity to tailor therapy to individual patients. With continual innovation, eventually real-time changes in treatment regimens can be made for individual patients to treat the ever-changing molecular landscape of the tumor. Immunotherapy remains the most promising treatment for advanced endometrial cancers.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 1","pages":"87 - 93"},"PeriodicalIF":1.2,"publicationDate":"2020-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1732204","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46020809","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: 2
Can we use disease burden to predict docetaxel activity in metastatic hormone-sensitive prostate cancer? 我们可以用疾病负担来预测转移性激素敏感前列腺癌的多西他赛活性吗?
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-02-12 DOI: 10.1080/23808993.2020.1727739
M. Randhawa, Robert J. Jones
One of the most important advances in the management of advanced prostate cancer in recent years, has been the utilization of drugs, previously reserved for those with metastatic castration-resista...
近年来晚期前列腺癌治疗中最重要的进展之一是药物的使用,这些药物以前只用于转移性去势抵抗性前列腺癌患者。
{"title":"Can we use disease burden to predict docetaxel activity in metastatic hormone-sensitive prostate cancer?","authors":"M. Randhawa, Robert J. Jones","doi":"10.1080/23808993.2020.1727739","DOIUrl":"https://doi.org/10.1080/23808993.2020.1727739","url":null,"abstract":"One of the most important advances in the management of advanced prostate cancer in recent years, has been the utilization of drugs, previously reserved for those with metastatic castration-resista...","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 1","pages":"55 - 57"},"PeriodicalIF":1.2,"publicationDate":"2020-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1727739","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46354649","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: 0
Moving beyond endocrine therapy for luminal metastatic breast cancer in the precision medicine era: looking for new targets 精准医疗时代癌症腔内转移性乳腺癌的内分泌治疗:寻找新靶点
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-01-02 DOI: 10.1080/23808993.2020.1720508
S. Morganti, G. Curigliano
ABSTRACT Introduction: Hormone receptor-positive (HR+) breast cancer (BC) is the most frequent BC subtype, for which estrogen receptor (ER)-signaling blockade still represents the cornerstone of treatments. Nevertheless, almost all HR+ BC patients develop resistance to endocrine therapy, a critical therapeutic unmet need for BC patients. Recent advances in genomic medicine allowed for a more detailed identification and better characterization of the mechanisms underlining endocrine resistance and led to the development of targeted agents potentially able to restore endocrine sensitivity, thus improving disease control and potentially survival. Areas covered: New drugs in the early phase of development for HR-positive metastatic BC (MBC) treatment are here outlined. PI3K/AKT/mTOR, FGFR, and IGFRs are the main pathways addressed, and evidences about HER2 blockade in triple-positive diseases are also reported. New hormonal agents active against estrogen receptor 1 (ESR1)-mutant MBC are presented. A brief mention of biological agents not targeting intracellular signaling hubs, such as histone-deacetylase inhibitors and immunotherapy drugs, is finally provided. Expert opinion: promising results have been reported for the majority of new drugs under development. Despite this, a ‘precision medicine approach’ still has to demonstrate a benefit in this disease. Identification of reliable predictive biomarkers should be a key objective of future researches.
摘要简介:激素受体阳性(HR+)癌症(BC)是最常见的BC亚型,雌激素受体(ER)信号传导阻断仍然是治疗的基石。然而,几乎所有HR+BC患者都对内分泌治疗产生耐药性,这是BC患者未满足的关键治疗需求。基因组医学的最新进展使人们能够更详细地识别和更好地表征内分泌抵抗的机制,并开发出可能恢复内分泌敏感性的靶向药物,从而改善疾病控制和潜在的生存率。涵盖的领域:这里概述了HR阳性转移性BC(MBC)治疗的早期开发新药。PI3K/AKT/mTOR、FGFR和IGFRs是解决的主要途径,也报道了HER2在三阳性疾病中阻断的证据。提出了新的抗雌激素受体1(ESR1)突变MBC的激素制剂。最后简要介绍了不靶向细胞内信号中枢的生物制剂,如组蛋白脱乙酰酶抑制剂和免疫疗法药物。专家意见:大多数正在开发的新药都报告了有希望的结果。尽管如此,“精准医学方法”仍然必须证明对这种疾病有好处。鉴定可靠的预测性生物标志物应该是未来研究的一个关键目标。
{"title":"Moving beyond endocrine therapy for luminal metastatic breast cancer in the precision medicine era: looking for new targets","authors":"S. Morganti, G. Curigliano","doi":"10.1080/23808993.2020.1720508","DOIUrl":"https://doi.org/10.1080/23808993.2020.1720508","url":null,"abstract":"ABSTRACT Introduction: Hormone receptor-positive (HR+) breast cancer (BC) is the most frequent BC subtype, for which estrogen receptor (ER)-signaling blockade still represents the cornerstone of treatments. Nevertheless, almost all HR+ BC patients develop resistance to endocrine therapy, a critical therapeutic unmet need for BC patients. Recent advances in genomic medicine allowed for a more detailed identification and better characterization of the mechanisms underlining endocrine resistance and led to the development of targeted agents potentially able to restore endocrine sensitivity, thus improving disease control and potentially survival. Areas covered: New drugs in the early phase of development for HR-positive metastatic BC (MBC) treatment are here outlined. PI3K/AKT/mTOR, FGFR, and IGFRs are the main pathways addressed, and evidences about HER2 blockade in triple-positive diseases are also reported. New hormonal agents active against estrogen receptor 1 (ESR1)-mutant MBC are presented. A brief mention of biological agents not targeting intracellular signaling hubs, such as histone-deacetylase inhibitors and immunotherapy drugs, is finally provided. Expert opinion: promising results have been reported for the majority of new drugs under development. Despite this, a ‘precision medicine approach’ still has to demonstrate a benefit in this disease. Identification of reliable predictive biomarkers should be a key objective of future researches.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"53 5","pages":"22 - 7"},"PeriodicalIF":1.2,"publicationDate":"2020-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1720508","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41296997","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: 4
Predicting therapeutic response through biomarker analysis in psoriatic arthritis, an example of precision medicine 通过生物标志物分析预测银屑病关节炎的治疗反应——精准医学的一个例子
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-01-02 DOI: 10.1080/23808993.2020.1724509
V. Chandran, P. Rahman
ABSTRACT Introduction: Psoriatic arthritis (PsA) is a heterogeneous inflammatory arthritis for which effective targeted therapies are now available. However, despite such advances up to 40% of patients fail to meet the primary endpoint in clinical trials. Current approach to therapy does not take it account clinical or molecular heterogeneity and is based on trial-and-error. It is envisioned that choosing therapies based on appropriate molecular biomarker profiles will help choose the appropriate therapy for a patient and as such lead to better treatment outcomes with reduced costs and less exposure to less effective therapies. Areas covered: The paper introduces PsA and briefly describes the clinical phenotype, pathogenesis, and current therapies. Current literature on biomarkers relating to PsA treatment response is reviewed. Limitations of the current approach, potential solutions to issues identified, and a path forward for research and potential clinical application in this area are discussed. Expert opinion: The likelihood that the current siloed approach to biomarker discovery will lead to meaningful clinically actionable tests to facilitate precision medicine in PsA is low. We envision that with collaborative and harmonized effort by all stakeholders using an inter-omic approach will lead to the development of robust predictive biomarkers for PsA treatment response.
摘要简介:银屑病关节炎(PsA)是一种异质性炎症性关节炎,目前已有有效的靶向治疗方法。然而,尽管取得了这些进展,仍有高达40%的患者未能达到临床试验的主要终点。目前的治疗方法没有考虑临床或分子异质性,而是基于反复试验。可以预见,基于适当的分子生物标志物图谱选择疗法将有助于为患者选择适当的疗法,并因此带来更好的治疗结果,同时降低成本,减少对低效疗法的接触。涵盖领域:本文介绍了PsA,并简要描述了其临床表型、发病机制和目前的治疗方法。综述了目前有关PsA治疗反应的生物标志物的文献。讨论了当前方法的局限性、已发现问题的潜在解决方案,以及该领域研究和潜在临床应用的前进道路。专家意见:目前孤立的生物标志物发现方法将导致有意义的临床可操作测试,以促进PsA的精准医学的可能性很低。我们设想,通过所有利益相关者的合作和协调努力,使用组间方法,将开发出用于PsA治疗反应的强大预测生物标志物。
{"title":"Predicting therapeutic response through biomarker analysis in psoriatic arthritis, an example of precision medicine","authors":"V. Chandran, P. Rahman","doi":"10.1080/23808993.2020.1724509","DOIUrl":"https://doi.org/10.1080/23808993.2020.1724509","url":null,"abstract":"ABSTRACT Introduction: Psoriatic arthritis (PsA) is a heterogeneous inflammatory arthritis for which effective targeted therapies are now available. However, despite such advances up to 40% of patients fail to meet the primary endpoint in clinical trials. Current approach to therapy does not take it account clinical or molecular heterogeneity and is based on trial-and-error. It is envisioned that choosing therapies based on appropriate molecular biomarker profiles will help choose the appropriate therapy for a patient and as such lead to better treatment outcomes with reduced costs and less exposure to less effective therapies. Areas covered: The paper introduces PsA and briefly describes the clinical phenotype, pathogenesis, and current therapies. Current literature on biomarkers relating to PsA treatment response is reviewed. Limitations of the current approach, potential solutions to issues identified, and a path forward for research and potential clinical application in this area are discussed. Expert opinion: The likelihood that the current siloed approach to biomarker discovery will lead to meaningful clinically actionable tests to facilitate precision medicine in PsA is low. We envision that with collaborative and harmonized effort by all stakeholders using an inter-omic approach will lead to the development of robust predictive biomarkers for PsA treatment response.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 1","pages":"35 - 42"},"PeriodicalIF":1.2,"publicationDate":"2020-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1724509","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42580454","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: 4
Challenges and advances in translating gene therapy for hearing disorders 翻译基因治疗听力障碍的挑战与进展
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-01-02 DOI: 10.1080/23808993.2020.1707077
H. Büning, A. Schambach, Michael A. Morgan, Axel Rossi, Helena Wichova, H. Staecker, A. Warnecke, T. Lenarz
ABSTRACT Introduction: Although sensorineural hearing loss (SNHL) is the most common neurodegenerative disease in humans, no approved pharmaceutical interventions are currently available. The progression of inherited as well as acquired forms of hearing loss can be altered by transferring single genes to distinct cell types of the ear inner. The inner ear is an attractive target for gene therapy given its small size and localized anatomic nature, which is accessible through routine surgical approaches. Areas covered: SNHL is the symptom of a diverse group of disorders with specific requirements regarding the timing of therapeutic intervention, the target cell population, the delivery system and, of course, the therapeutic active “substance’. Expert opinion: Despite these challenges, which will be discussed in the following paper, the first human gene therapy clinical trial for an inner ear disease is already initiated, making this the perfect time to translate a great variety of therapeutic approaches from the laboratory into clinical routine.
摘要简介:尽管感觉神经性听力损失(SNHL)是人类最常见的神经退行性疾病,但目前还没有批准的药物干预措施。遗传性和后天性听力损失的进展可以通过将单个基因转移到不同类型的内耳细胞来改变。内耳是基因治疗的一个有吸引力的靶点,因为它的体积小,解剖结构局限,可以通过常规手术进入。涵盖领域:SNHL是一组不同疾病的症状,对治疗干预的时间、靶细胞群、递送系统,当然还有,治疗活性“物质”。专家意见:尽管存在这些挑战(将在下面的论文中讨论),但第一项针对内耳疾病的人类基因治疗临床试验已经启动,这是将实验室的各种治疗方法转化为临床常规的最佳时机。
{"title":"Challenges and advances in translating gene therapy for hearing disorders","authors":"H. Büning, A. Schambach, Michael A. Morgan, Axel Rossi, Helena Wichova, H. Staecker, A. Warnecke, T. Lenarz","doi":"10.1080/23808993.2020.1707077","DOIUrl":"https://doi.org/10.1080/23808993.2020.1707077","url":null,"abstract":"ABSTRACT Introduction: Although sensorineural hearing loss (SNHL) is the most common neurodegenerative disease in humans, no approved pharmaceutical interventions are currently available. The progression of inherited as well as acquired forms of hearing loss can be altered by transferring single genes to distinct cell types of the ear inner. The inner ear is an attractive target for gene therapy given its small size and localized anatomic nature, which is accessible through routine surgical approaches. Areas covered: SNHL is the symptom of a diverse group of disorders with specific requirements regarding the timing of therapeutic intervention, the target cell population, the delivery system and, of course, the therapeutic active “substance’. Expert opinion: Despite these challenges, which will be discussed in the following paper, the first human gene therapy clinical trial for an inner ear disease is already initiated, making this the perfect time to translate a great variety of therapeutic approaches from the laboratory into clinical routine.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 1","pages":"23 - 34"},"PeriodicalIF":1.2,"publicationDate":"2020-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1707077","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43853547","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: 1
Patient-derived organoid analysis of drug resistance in precision medicine: is there a value? 精准医学患者来源类器官耐药分析:有价值吗?
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-01-02 DOI: 10.1080/23808993.2020.1715794
Amira Abugomaa, M. Elbadawy
Cancer is the main health problem with high morbidity and mortality. The worldwide annual incidence and deaths from cancers are estimated in millions [1,2]. Hence, cancer therapy must be continuously promoted and developed. Cancer study, like in other diseases, highly depends on representative and reliable models. However, the tumor is not uniform, but rather a heterogenic and highly variable and complex than other diseases, rendering its study extremely difficult, expensive [3]. The most common treatment methods for cancers are based on surgery [4], chemotherapy [5], radiotherapy [6], and immunotherapy [7]. The response of cancers to these various treatment strategies differs according to tumor subtype, clinical stage, and associated risk factors and unfortunately fails to limit the progression of cancer in various cases. Even the same tumor of the same organ or tissue differs in response to therapy among patients and the recurrence and metastasis which are associated with high resistance to therapy are main issues [8]. Also, chemotherapy affects the quality of life because of its potential side effects and therefore disfavored by many patients. Therefore, suitable models to expect the treatment response with high precision are of extreme need toward more personalized treatment of patients. Experimental models are assigned to understand the pathobiology, identify diagnostic and prognostic biomarkers of cancer progression and establish novel potent and effective therapies. To date, the available treatment protocols seem inadequate to prevent the resistance to therapy as well as the recurrence and progression of cancer. Therefore, several experimental models of to study cancer development and progression including rats and mice as well as the in vitro culture models of cell line, 2D cell, induced pluripotent stem cell (iPSCs) lines, spheroids, 3D organoids, organotypic tissue slice cultures, patient-derived tumor xenografts are developed [9–11] and certainly, they confer valuable tools for preclinical pharmacological assessment. Cancer stem cells (CSCs) are tumor-initiating cells capable of conserving cellular heterogeneity, self-renewal, epithelial to mesenchymal transition, differentiation to form all kind cells in a given tumor [12] and drive the tumor growth, metastasis and most importantly, the resistance to conventional anticancer agents [13–22]. Therefore, the total eradication of CSCs is crucial for the successful treatment of cancers. Thus, understanding the mechanism of response and resistance of CSCs to therapy and developing personalized therapy is the cornerstone for treatments of cancer in cancer patients. Currently, the selection of the correct tool to use in the laboratory to elucidate the mechanism of CSCs’ resistance to therapy can depend on the question on hand, but also on the resources (and knowledge) available. Among them, organoids constitute the more reasonable method in recapitulating the in vivo microenvironment of the tu
癌症是主要的健康问题,发病率和死亡率都很高。据估计,全球每年癌症的发病率和死亡人数以百万计[1,2]。因此,癌症治疗必须不断地推广和发展。癌症研究和其他疾病一样,高度依赖于具有代表性和可靠性的模型。然而,与其他疾病相比,肿瘤并不均匀,而是一种异质性、高度可变和复杂的疾病,这使得其研究极其困难和昂贵[3]。癌症最常见的治疗方法是基于手术[4]、化疗[5]、放疗[6]和免疫疗法[7]。癌症对这些不同治疗策略的反应因肿瘤亚型、临床分期和相关风险因素而异,不幸的是,在各种情况下,未能限制癌症的进展。即使是同一器官或组织的同一肿瘤,患者对治疗的反应也不同,与高耐药性相关的复发和转移是主要问题[8]。此外,化疗由于其潜在的副作用而影响生活质量,因此不受许多患者的欢迎。因此,对于更个性化的患者治疗,迫切需要合适的模型来期望高精度的治疗反应。实验模型被分配来理解病理生物学,识别癌症进展的诊断和预后生物标志物,并建立新的有效疗法。到目前为止,现有的治疗方案似乎不足以防止对治疗的耐药性以及癌症的复发和进展。因此,开发了几种研究癌症发展和进展的实验模型,包括大鼠和小鼠,以及细胞系、2D细胞、诱导多能干细胞(iPSC)系、球体、3D类器官、器官型组织切片培养物、患者来源的肿瘤异种移植物的体外培养模型[9-11],当然,它们为临床前药理学评估提供了宝贵的工具。癌症干细胞(CSCs)是肿瘤起始细胞,能够保存细胞异质性、自我更新、上皮向间充质转化、分化以在特定肿瘤中形成所有类型的细胞[12],并驱动肿瘤生长、转移,最重要的是,驱动对传统抗癌药物的耐药性[13-22]。因此,彻底根除CSCs对于成功治疗癌症至关重要。因此,了解CSC对治疗的反应和抵抗机制,开发个性化治疗是癌症患者治疗癌症的基石。目前,在实验室中选择正确的工具来阐明CSC对治疗的抵抗机制可能取决于手头的问题,也取决于可用的资源(和知识)。其中,类器官构成了更合理的方法来概括肿瘤的体内微环境,并且比切片培养或异种移植物更容易处理和高通量筛选。在过去的十年里,研究人员在类器官生物学领域取得了实质性进展,其他地方已经对类器官系统进行了详细的综述[21,23,24]。
{"title":"Patient-derived organoid analysis of drug resistance in precision medicine: is there a value?","authors":"Amira Abugomaa, M. Elbadawy","doi":"10.1080/23808993.2020.1715794","DOIUrl":"https://doi.org/10.1080/23808993.2020.1715794","url":null,"abstract":"Cancer is the main health problem with high morbidity and mortality. The worldwide annual incidence and deaths from cancers are estimated in millions [1,2]. Hence, cancer therapy must be continuously promoted and developed. Cancer study, like in other diseases, highly depends on representative and reliable models. However, the tumor is not uniform, but rather a heterogenic and highly variable and complex than other diseases, rendering its study extremely difficult, expensive [3]. The most common treatment methods for cancers are based on surgery [4], chemotherapy [5], radiotherapy [6], and immunotherapy [7]. The response of cancers to these various treatment strategies differs according to tumor subtype, clinical stage, and associated risk factors and unfortunately fails to limit the progression of cancer in various cases. Even the same tumor of the same organ or tissue differs in response to therapy among patients and the recurrence and metastasis which are associated with high resistance to therapy are main issues [8]. Also, chemotherapy affects the quality of life because of its potential side effects and therefore disfavored by many patients. Therefore, suitable models to expect the treatment response with high precision are of extreme need toward more personalized treatment of patients. Experimental models are assigned to understand the pathobiology, identify diagnostic and prognostic biomarkers of cancer progression and establish novel potent and effective therapies. To date, the available treatment protocols seem inadequate to prevent the resistance to therapy as well as the recurrence and progression of cancer. Therefore, several experimental models of to study cancer development and progression including rats and mice as well as the in vitro culture models of cell line, 2D cell, induced pluripotent stem cell (iPSCs) lines, spheroids, 3D organoids, organotypic tissue slice cultures, patient-derived tumor xenografts are developed [9–11] and certainly, they confer valuable tools for preclinical pharmacological assessment. Cancer stem cells (CSCs) are tumor-initiating cells capable of conserving cellular heterogeneity, self-renewal, epithelial to mesenchymal transition, differentiation to form all kind cells in a given tumor [12] and drive the tumor growth, metastasis and most importantly, the resistance to conventional anticancer agents [13–22]. Therefore, the total eradication of CSCs is crucial for the successful treatment of cancers. Thus, understanding the mechanism of response and resistance of CSCs to therapy and developing personalized therapy is the cornerstone for treatments of cancer in cancer patients. Currently, the selection of the correct tool to use in the laboratory to elucidate the mechanism of CSCs’ resistance to therapy can depend on the question on hand, but also on the resources (and knowledge) available. Among them, organoids constitute the more reasonable method in recapitulating the in vivo microenvironment of the tu","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 1","pages":"1 - 5"},"PeriodicalIF":1.2,"publicationDate":"2020-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1715794","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44336799","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: 26
Roles of CRAC channel in cancer: implications for therapeutic development. CRAC通道在癌症中的作用:对治疗发展的影响。
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-01-01 Epub Date: 2020-08-11 DOI: 10.1080/23808993.2020.1803062
Husain Yar Khan, Iqra Mazahir, Shriya Reddy, Farzeen Fazili, AsfarSohail Azmi

Introduction: The Ca2+release-activated Ca2+ (CRAC) channel, composed of Orai and STIM proteins, represents one of the main routes of Ca2+ entry in most non-excitable cells. There is accumulating evidence to suggest that CRAC channel can influence various processes associated with tumorigenesis. Overexpression of CRAC channel proteins has been observed in several types of cancer tissues and cells, indicating that blocking CRAC channel activated Ca2+ influx can have therapeutic benefits for cancer patients.

Areas covered: In this review, we have primarily focused on the molecular composition and activation mechanism of CRAC channel as well as the myriad roles this Ca2+ channel play in various cancers. We further describe relevant information about several efforts aimed at developing CRAC channel blockers and their likely implications for cancer therapy. We have extensively utilized the available literature on PubMed to this end.

Expert opinion: The possibility of targeting CRAC channel mediated Ca2+ entry in cancer cells has generated considerable interest in recent years. Use of CRAC channel blockers in cancer preclinical studies and clinical trials has been relatively limited as compared to other diseases. The future lies in developing and testing more potent and selective drugs that target cancer cell specific CRAC channel proteins, hence opening better avenues for cancer therapeutic development.

Ca2+释放激活的Ca2+ (CRAC)通道由Orai和STIM蛋白组成,是大多数不可兴奋细胞中Ca2+进入的主要途径之一。越来越多的证据表明,CRAC通道可以影响与肿瘤发生相关的各种过程。在几种类型的癌症组织和细胞中已经观察到CRAC通道蛋白的过表达,这表明阻断CRAC通道激活的Ca2+内流可以对癌症患者具有治疗益处。研究领域:本文主要研究了钙离子通道的分子组成和激活机制,以及钙离子通道在多种癌症中的作用。我们进一步描述了一些旨在开发CRAC通道阻滞剂的相关信息及其对癌症治疗的可能影响。为此,我们广泛地利用了PubMed上的可用文献。专家意见:靶向癌细胞中CRAC通道介导的Ca2+进入的可能性近年来引起了相当大的兴趣。与其他疾病相比,CRAC通道阻滞剂在癌症临床前研究和临床试验中的应用相对有限。未来在于开发和测试针对癌细胞特异性CRAC通道蛋白的更有效和选择性的药物,从而为癌症治疗开发开辟更好的途径。
{"title":"Roles of CRAC channel in cancer: implications for therapeutic development.","authors":"Husain Yar Khan,&nbsp;Iqra Mazahir,&nbsp;Shriya Reddy,&nbsp;Farzeen Fazili,&nbsp;AsfarSohail Azmi","doi":"10.1080/23808993.2020.1803062","DOIUrl":"https://doi.org/10.1080/23808993.2020.1803062","url":null,"abstract":"<p><strong>Introduction: </strong>The Ca2+release-activated Ca2+ (CRAC) channel, composed of Orai and STIM proteins, represents one of the main routes of Ca<sup>2+</sup> entry in most non-excitable cells. There is accumulating evidence to suggest that CRAC channel can influence various processes associated with tumorigenesis. Overexpression of CRAC channel proteins has been observed in several types of cancer tissues and cells, indicating that blocking CRAC channel activated Ca<sup>2+</sup> influx can have therapeutic benefits for cancer patients.</p><p><strong>Areas covered: </strong>In this review, we have primarily focused on the molecular composition and activation mechanism of CRAC channel as well as the myriad roles this Ca<sup>2+</sup> channel play in various cancers. We further describe relevant information about several efforts aimed at developing CRAC channel blockers and their likely implications for cancer therapy. We have extensively utilized the available literature on PubMed to this end.</p><p><strong>Expert opinion: </strong>The possibility of targeting CRAC channel mediated Ca<sup>2+</sup> entry in cancer cells has generated considerable interest in recent years. Use of CRAC channel blockers in cancer preclinical studies and clinical trials has been relatively limited as compared to other diseases. The future lies in developing and testing more potent and selective drugs that target cancer cell specific CRAC channel proteins, hence opening better avenues for cancer therapeutic development.</p>","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 5","pages":"371-382"},"PeriodicalIF":1.2,"publicationDate":"2020-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1803062","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"25485856","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: 2
Proteomics-inspired precision medicine for treating and understanding multiple myeloma. 蛋白质组学启发的精准医学治疗和理解多发性骨髓瘤。
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2020-01-01 Epub Date: 2020-02-24 DOI: 10.1080/23808993.2020.1732205
Matthew Ho, Giada Bianchi, Kenneth C Anderson

Introduction: Remarkable progress in molecular characterization methods has led to significant improvements in how we manage multiple myeloma (MM). The introduction of novel therapies has led to significant improvements in overall survival over the past 10 years. However, MM remains incurable and treatment choice is largely based on outdated risk-adaptive strategies that do not factor in improved treatment outcomes in the context of modern therapies.

Areas covered: This review discusses current risk-adaptive strategies in MM and the clinical application of proteomics in the monitoring of treatment response, disease progression, and minimal residual disease (MRD). We also discuss promising biomarkers of disease progression, treatment response, and chemoresistance. Finally, we will discuss an immunomics-based approach to monoclonal antibody (mAb), vaccine, and CAR-T cell development.

Expert opinion: It is an exciting era in oncology with basic scientific knowledge translating in novel therapeutic approaches to improve patient outcomes. With the advent of effective immunotherapies and targeted therapies, it has become crucial to identify biomarkers to aid in the stratification of patients based on anticipated sensitivity to chemotherapy. As a paradigm of diseases highly dependent on protein homeostasis, multiple myeloma provides the perfect opportunity to investigate the use of proteomics to aid in precision medicine.

简介:分子表征方法的显著进步导致了我们如何管理多发性骨髓瘤(MM)的显着改进。在过去的10年里,新疗法的引入导致了总体生存率的显著提高。然而,多发性骨髓瘤仍然无法治愈,治疗选择主要基于过时的风险适应策略,在现代治疗背景下,这些策略没有考虑到改善的治疗结果。涵盖领域:本综述讨论了目前MM的风险适应策略以及蛋白质组学在监测治疗反应、疾病进展和最小残留病(MRD)中的临床应用。我们还讨论了有希望的疾病进展、治疗反应和化疗耐药的生物标志物。最后,我们将讨论基于免疫组学的单克隆抗体(mAb)、疫苗和CAR-T细胞发育方法。专家意见:这是肿瘤学的一个激动人心的时代,基础科学知识转化为新的治疗方法,以改善患者的预后。随着有效的免疫疗法和靶向治疗的出现,识别生物标志物以帮助基于化疗预期敏感性的患者分层变得至关重要。作为一种高度依赖蛋白质稳态的疾病范例,多发性骨髓瘤为研究蛋白质组学在精准医学中的应用提供了绝佳的机会。
{"title":"Proteomics-inspired precision medicine for treating and understanding multiple myeloma.","authors":"Matthew Ho,&nbsp;Giada Bianchi,&nbsp;Kenneth C Anderson","doi":"10.1080/23808993.2020.1732205","DOIUrl":"https://doi.org/10.1080/23808993.2020.1732205","url":null,"abstract":"<p><strong>Introduction: </strong>Remarkable progress in molecular characterization methods has led to significant improvements in how we manage multiple myeloma (MM). The introduction of novel therapies has led to significant improvements in overall survival over the past 10 years. However, MM remains incurable and treatment choice is largely based on outdated risk-adaptive strategies that do not factor in improved treatment outcomes in the context of modern therapies.</p><p><strong>Areas covered: </strong>This review discusses current risk-adaptive strategies in MM and the clinical application of proteomics in the monitoring of treatment response, disease progression, and minimal residual disease (MRD). We also discuss promising biomarkers of disease progression, treatment response, and chemoresistance. Finally, we will discuss an immunomics-based approach to monoclonal antibody (mAb), vaccine, and CAR-T cell development.</p><p><strong>Expert opinion: </strong>It is an exciting era in oncology with basic scientific knowledge translating in novel therapeutic approaches to improve patient outcomes. With the advent of effective immunotherapies and targeted therapies, it has become crucial to identify biomarkers to aid in the stratification of patients based on anticipated sensitivity to chemotherapy. As a paradigm of diseases highly dependent on protein homeostasis, multiple myeloma provides the perfect opportunity to investigate the use of proteomics to aid in precision medicine.</p>","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"5 2","pages":"67-85"},"PeriodicalIF":1.2,"publicationDate":"2020-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2020.1732205","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39330829","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: 5
期刊
Expert Review of Precision Medicine and Drug Development
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