首页 > 最新文献

Expert Review of Precision Medicine and Drug Development最新文献

英文 中文
Genotype-Guided vs Clinically-Guided Stable Warfarin Dose Prediction and Stable Dose Establishment In A Predominantly Non-European Ancestry Population 基因型引导与临床引导的主要非欧洲祖先群体中华法林稳定剂量预测和稳定剂量建立
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-10-28 DOI: 10.1080/23808993.2021.1989303
Annesti F. Elmasri, Hee-soo Hur, Jin Han, James C. Lee
ABSTRACT Background Warfarin dosing varies due to individual genetic and clinical factors. The utility of genotype‐guided warfarin dosing to improve stable warfarin dose is not extensively studied in non-European populations. Research design and methods Retrospective cohort study of patients initiating warfarin receiving genotype (PGx)‐guided or clinically guided dosing. Primary outcomes included dose discordance between estimated dose at discharge and eventual stable dose. Results No significant difference in warfarin dose discordance was observed (PGx: 9.1 ± 8.8 mg/week difference vs. Clinical: 7.9 ± 8.9 mg/week difference; P = 0.446). PGx‐guided dosing did not reduce time to achieve stable dose (1.8 ± 2.5 months vs. 2.1 ± 2.6 months; P = 0.508). Vitamin K intake level did not alter prediction accuracy or time to stable dose (PGx‐predicted: dose difference P = 0.493, time to stable dose P = 0.336; Clinically predicted: dose difference P = 0.145, time to dose INR P = 0.095). Conclusions PGx‐guided warfarin dosing did not improve eventual stable dose discordance or reduce the time to achieve stable dose in this predominantly non‐European cohort. Dietary vitamin K intake level did not impact warfarin dose discordance or time to achieve stable warfarin dose. Additional study is needed to identify populations that best benefit from PGx‐guided warfarin dosing.
背景华法林的剂量因个体遗传和临床因素而异。在非欧洲人群中,基因型引导华法林剂量提高华法林稳定剂量的效用尚未得到广泛研究。研究设计和方法对接受基因型(PGx)指导或临床指导给药的华法林患者进行回顾性队列研究。主要结局包括出院时估计剂量与最终稳定剂量之间的剂量不一致。结果两组华法林剂量差异无显著性差异(PGx: 9.1±8.8 mg/周差异与临床:7.9±8.9 mg/周差异;P = 0.446)。PGx引导给药并没有缩短达到稳定剂量的时间(1.8±2.5个月vs. 2.1±2.6个月);P = 0.508)。维生素K摄入水平不改变预测准确性或达到稳定剂量所需时间(PGx‐预测:剂量差P = 0.493,达到稳定剂量所需时间P = 0.336;临床预测:剂量差P = 0.145,至剂量时间INR P = 0.095)。结论:PGx引导的华法林给药并没有改善最终的稳定剂量不一致或减少达到稳定剂量的时间。膳食维生素K摄入量水平不影响华法林剂量不一致或达到稳定华法林剂量的时间。需要进一步的研究来确定从PGx引导的华法林剂量中获益最大的人群。
{"title":"Genotype-Guided vs Clinically-Guided Stable Warfarin Dose Prediction and Stable Dose Establishment In A Predominantly Non-European Ancestry Population","authors":"Annesti F. Elmasri, Hee-soo Hur, Jin Han, James C. Lee","doi":"10.1080/23808993.2021.1989303","DOIUrl":"https://doi.org/10.1080/23808993.2021.1989303","url":null,"abstract":"ABSTRACT Background Warfarin dosing varies due to individual genetic and clinical factors. The utility of genotype‐guided warfarin dosing to improve stable warfarin dose is not extensively studied in non-European populations. Research design and methods Retrospective cohort study of patients initiating warfarin receiving genotype (PGx)‐guided or clinically guided dosing. Primary outcomes included dose discordance between estimated dose at discharge and eventual stable dose. Results No significant difference in warfarin dose discordance was observed (PGx: 9.1 ± 8.8 mg/week difference vs. Clinical: 7.9 ± 8.9 mg/week difference; P = 0.446). PGx‐guided dosing did not reduce time to achieve stable dose (1.8 ± 2.5 months vs. 2.1 ± 2.6 months; P = 0.508). Vitamin K intake level did not alter prediction accuracy or time to stable dose (PGx‐predicted: dose difference P = 0.493, time to stable dose P = 0.336; Clinically predicted: dose difference P = 0.145, time to dose INR P = 0.095). Conclusions PGx‐guided warfarin dosing did not improve eventual stable dose discordance or reduce the time to achieve stable dose in this predominantly non‐European cohort. Dietary vitamin K intake level did not impact warfarin dose discordance or time to achieve stable warfarin dose. Additional study is needed to identify populations that best benefit from PGx‐guided warfarin dosing.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"375 - 379"},"PeriodicalIF":1.2,"publicationDate":"2021-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42981298","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
Atezolizumab plus vemurafenib and cobimetinib for the treatment of BRAF V600-mutant advanced melanoma: from an hypothetic triplet to an approved regimen 阿替佐利珠单抗联合维穆拉非尼和科比替尼治疗BRAF V600突变晚期黑色素瘤:从假设的三重方案到批准的方案
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-09-20 DOI: 10.1080/23808993.2021.1976637
Aikaterini Katsandris, D. Ziogas, M. Kontouri, Stavroula Staikoglou, H. Gogas
ABSTRACT Introduction Breakthrough changes in melanoma management induced by the introduction of BRAF/MEK inhibitors and immune checkpoint inhibitors (ICPIs) have motivated subsequent research to combine these strategies in the frontline approach of BRAFV600-mutant advanced melanoma. Initial preclinical data have shown that targeted agents may have a favorable immune impact on the tumor microenvironment and next, numerous trials tried to identify the optimal combination and sequence of immunotherapy and targeted therapy. Up until last year, when the first immune/targeted regimen including atezolizumab, vemurafenib and cobimetinib, was approved by the FDA. Areas covered Focusing on this first-in-class immune/targeted regimen, we describe here the entire process for its approval, from the bench of preclinical studies to the clinical evaluation of its efficacy and its toxicity profile on treated patients with advanced BRAF-mutant melanoma. All raised concerns about the use of atezolizumab/vemurafenib/cobimetinib triplet are thoroughly discussed. Expert opinion The atezolizumab/vemurafenib/cobimetinib triplet is an effective frontline option for advanced BRAF-mutant melanoma but its generalized implementation remains limited due to toxicity and cost. Results of ongoing trials are expected to identify more clearly the best candidates for such combinations and to update the role of immune/targeted triplets in melanoma treatment.
摘要简介BRAF/MEK抑制剂和免疫检查点抑制剂(ICPI)的引入引发了黑色素瘤管理的突破性变化,促使随后的研究将这些策略结合到BRAFV600突变晚期黑色素瘤的一线方法中。最初的临床前数据表明,靶向药物可能对肿瘤微环境产生有利的免疫影响,接下来,许多试验试图确定免疫疗法和靶向治疗的最佳组合和顺序。直到去年,包括atezolizumab、vemurafenib和cobimetinib在内的第一个免疫/靶向方案获得了美国食品药品监督管理局的批准。涵盖的领域专注于这一一流的免疫/靶向方案,我们在这里描述了其批准的整个过程,从临床前研究到对其治疗晚期BRAF突变黑色素瘤患者的疗效和毒性的临床评估。对atezolizumab/vemurafenib/cobimetinib三联用药引起的所有关注进行了彻底讨论。专家意见atezolizumab/vemurafenib/cobimetinib三联用药是治疗晚期BRAF突变黑色素瘤的有效一线选择,但由于毒性和成本原因,其广泛应用仍然有限。正在进行的试验结果有望更清楚地确定这种组合的最佳候选者,并更新免疫/靶向三联体在黑色素瘤治疗中的作用。
{"title":"Atezolizumab plus vemurafenib and cobimetinib for the treatment of BRAF V600-mutant advanced melanoma: from an hypothetic triplet to an approved regimen","authors":"Aikaterini Katsandris, D. Ziogas, M. Kontouri, Stavroula Staikoglou, H. Gogas","doi":"10.1080/23808993.2021.1976637","DOIUrl":"https://doi.org/10.1080/23808993.2021.1976637","url":null,"abstract":"ABSTRACT Introduction Breakthrough changes in melanoma management induced by the introduction of BRAF/MEK inhibitors and immune checkpoint inhibitors (ICPIs) have motivated subsequent research to combine these strategies in the frontline approach of BRAFV600-mutant advanced melanoma. Initial preclinical data have shown that targeted agents may have a favorable immune impact on the tumor microenvironment and next, numerous trials tried to identify the optimal combination and sequence of immunotherapy and targeted therapy. Up until last year, when the first immune/targeted regimen including atezolizumab, vemurafenib and cobimetinib, was approved by the FDA. Areas covered Focusing on this first-in-class immune/targeted regimen, we describe here the entire process for its approval, from the bench of preclinical studies to the clinical evaluation of its efficacy and its toxicity profile on treated patients with advanced BRAF-mutant melanoma. All raised concerns about the use of atezolizumab/vemurafenib/cobimetinib triplet are thoroughly discussed. Expert opinion The atezolizumab/vemurafenib/cobimetinib triplet is an effective frontline option for advanced BRAF-mutant melanoma but its generalized implementation remains limited due to toxicity and cost. Results of ongoing trials are expected to identify more clearly the best candidates for such combinations and to update the role of immune/targeted triplets in melanoma treatment.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"349 - 360"},"PeriodicalIF":1.2,"publicationDate":"2021-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49442913","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
Individualized management of cytomegalovirus in solid organ transplant recipients 实体器官移植受者巨细胞病毒的个体化治疗
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-09-03 DOI: 10.1080/23808993.2021.1964951
H. Saeed, Matthew J. Thoendel, R. Razonable
ABSTRACT Introduction Cytomegalovirus (CMV) is an opportunistic infection that affects immunocompromised solid organ transplant patients. Defining the imbalance between host and virus factors that predispose to its occurrence can assist in individualizing the approach to CMV prevention and treatment. Areas covered In this narrative review article, we provide an up to date overview of host, pathogen, and transplant-related factors that determine the risk and outcome of CMV infection in solid organ transplant recipients. We review the role of CMV-specific cell-mediated and humoral immune status, degree of lymphopenia, degree of viremia, and the dose and type of immunosuppressive regimen in defining the risk and determining the outcome of CMV. We propose that knowledge of these factors should be taken into account in optimizing the management strategies and individualize our approach to CMV prevention and treatment in the posttransplant setting. Expert opinion The management of CMV in transplant recipients is not a one-size-fits-all strategy. We highlight the spectrum of CMV risk and outcomes in solid organ transplant recipients based on host, virus, and transplant-related factors. We provide examples on how to incorporate these factors in the implementation of optimized antiviral prophylaxis, preemptive treatment of asymptomatic infection and management of susceptible, refractory, and resistant CMV disease.
巨细胞病毒(CMV)是一种影响免疫功能低下的实体器官移植患者的机会性感染。确定易导致其发生的宿主和病毒因素之间的不平衡有助于个体化CMV预防和治疗方法。在这篇叙述性综述文章中,我们提供了宿主、病原体和移植相关因素的最新综述,这些因素决定了实体器官移植受者巨细胞病毒感染的风险和结果。我们回顾了CMV特异性细胞介导和体液免疫状态、淋巴细胞减少程度、病毒血症程度以及免疫抑制方案的剂量和类型在确定CMV风险和决定结果中的作用。我们建议,在优化管理策略和个性化移植后CMV预防和治疗方法时,应考虑到这些因素。专家意见移植受者巨细胞病毒的管理并不是一个放之四海而皆准的策略。我们强调基于宿主、病毒和移植相关因素的实体器官移植受者巨细胞病毒风险和结果的谱。我们提供了如何将这些因素纳入实施优化的抗病毒预防,无症状感染的先发制人治疗和易感,难治性和耐药巨细胞病毒疾病的管理的例子。
{"title":"Individualized management of cytomegalovirus in solid organ transplant recipients","authors":"H. Saeed, Matthew J. Thoendel, R. Razonable","doi":"10.1080/23808993.2021.1964951","DOIUrl":"https://doi.org/10.1080/23808993.2021.1964951","url":null,"abstract":"ABSTRACT Introduction Cytomegalovirus (CMV) is an opportunistic infection that affects immunocompromised solid organ transplant patients. Defining the imbalance between host and virus factors that predispose to its occurrence can assist in individualizing the approach to CMV prevention and treatment. Areas covered In this narrative review article, we provide an up to date overview of host, pathogen, and transplant-related factors that determine the risk and outcome of CMV infection in solid organ transplant recipients. We review the role of CMV-specific cell-mediated and humoral immune status, degree of lymphopenia, degree of viremia, and the dose and type of immunosuppressive regimen in defining the risk and determining the outcome of CMV. We propose that knowledge of these factors should be taken into account in optimizing the management strategies and individualize our approach to CMV prevention and treatment in the posttransplant setting. Expert opinion The management of CMV in transplant recipients is not a one-size-fits-all strategy. We highlight the spectrum of CMV risk and outcomes in solid organ transplant recipients based on host, virus, and transplant-related factors. We provide examples on how to incorporate these factors in the implementation of optimized antiviral prophylaxis, preemptive treatment of asymptomatic infection and management of susceptible, refractory, and resistant CMV disease.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"333 - 344"},"PeriodicalIF":1.2,"publicationDate":"2021-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44354255","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
Targeted immunotherapies to consider for B Cell non-hodgkin lymphoma B细胞非霍奇金淋巴瘤的靶向免疫治疗
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-09-03 DOI: 10.1080/23808993.2021.1967142
E. Namoglu, M. Hughes, S. Nasta
ABSTRACT Introduction Non-Hodgkin lymphoma is a disease spectrum of multiple subtypes with many potential targets. Given the heterogeneity and evolving landscape for targeted therapy, incorporation of therapeutics in this disease state is complex and dependent upon multiple factors. Areas covered Agents reviewed for either on- or off-label use include: monoclonal antibodies, antibody-drug conjugates, immunotherapy, cellular therapy, and bi-specific antibodies. In the current review, we discuss the most recently identified targets of interest and corresponding therapies. The authors aim to provide insight on where therapies may be incorporated for clinical utility in B cell non-Hodgkin lymphoma as well as future directions for new targets and combinations of these approaches. Expert opinion A multimodality approach to non-Hodgkin lymphoma will now be needed for early identification of potential targets and will be critical for treatment decisions. As each agent is defined in the relapsed refractory setting, the sequencing and combination of these agents will be critical, particularly with overlapping toxicities. The adoption in practice will depend not only on efficacy but ease of administration, limiting side effects and cost.
非霍奇金淋巴瘤是一种多亚型的疾病,具有许多潜在的靶点。鉴于靶向治疗的异质性和不断发展的前景,在这种疾病状态下结合治疗是复杂的,取决于多种因素。在说明书上或说明书外使用的药物包括:单克隆抗体、抗体-药物偶联物、免疫治疗、细胞治疗和双特异性抗体。在当前的综述中,我们讨论了最近发现的感兴趣的靶点和相应的治疗方法。作者旨在为B细胞非霍奇金淋巴瘤的临床应用提供见解,以及新靶点和这些方法组合的未来方向。现在需要采用多模式治疗非霍奇金淋巴瘤,以早期识别潜在靶点,并将对治疗决策至关重要。由于每种药物都是在复发的难治性环境中定义的,因此这些药物的排序和组合将是至关重要的,特别是在重叠毒性的情况下。在实践中采用不仅取决于疗效,而且取决于是否易于使用、限制副作用和费用。
{"title":"Targeted immunotherapies to consider for B Cell non-hodgkin lymphoma","authors":"E. Namoglu, M. Hughes, S. Nasta","doi":"10.1080/23808993.2021.1967142","DOIUrl":"https://doi.org/10.1080/23808993.2021.1967142","url":null,"abstract":"ABSTRACT Introduction Non-Hodgkin lymphoma is a disease spectrum of multiple subtypes with many potential targets. Given the heterogeneity and evolving landscape for targeted therapy, incorporation of therapeutics in this disease state is complex and dependent upon multiple factors. Areas covered Agents reviewed for either on- or off-label use include: monoclonal antibodies, antibody-drug conjugates, immunotherapy, cellular therapy, and bi-specific antibodies. In the current review, we discuss the most recently identified targets of interest and corresponding therapies. The authors aim to provide insight on where therapies may be incorporated for clinical utility in B cell non-Hodgkin lymphoma as well as future directions for new targets and combinations of these approaches. Expert opinion A multimodality approach to non-Hodgkin lymphoma will now be needed for early identification of potential targets and will be critical for treatment decisions. As each agent is defined in the relapsed refractory setting, the sequencing and combination of these agents will be critical, particularly with overlapping toxicities. The adoption in practice will depend not only on efficacy but ease of administration, limiting side effects and cost.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"317 - 332"},"PeriodicalIF":1.2,"publicationDate":"2021-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46060593","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
Recent advances in precision medicine for the treatment of medullary thyroid cancer 精准医学治疗甲状腺髓样癌的最新进展
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-09-03 DOI: 10.1080/23808993.2021.1964952
J. Krajewska, A. Kukulska, M. Oczko-Wojciechowska, B. Jarzab
ABSTRACT Introduction The discovery of a pivotal role of tyrosine kinases in the pathogenesis of medullary thyroid carcinoma (MTC) opened up new options in the systemic treatment of advanced disease. During the last decade, two multikinase inhibitors (MKIs) – vandetanib and cabozantinib and, more recently, two potent, selective RET inhibitors selpercatinib and pralsetinib gained regulatory approval. Areas covered The data on efficacy and safety of vandetanib, cabozantinib, selpercatinib, and pralsetinib in MTC published during the recent 10 years. Expert opinion The perspectives of systemic MTC treatment have substantially changed since 2010. However, to date, the question of which drug should be chosen as the first line remains open. There are no recommendations on what to use as the second or other treatment lines. The impact of tumor burden, progression slope, and the presence of MTC symptoms on treatment-decision making is not unequivocally defined. Another important problem is the treatment duration. EBM (evidence-based medicine) study, resolving these issues, is our task for the nearest future. The treatment tolerability and its impact on the quality of life, particularly regarding nonselective MKIs, is also an essential problem.
酪氨酸激酶在甲状腺髓样癌(MTC)发病机制中的关键作用的发现为晚期疾病的全身治疗开辟了新的选择。在过去的十年中,两种多激酶抑制剂(MKIs) - vandetanib和cabozantinib,以及最近两种有效的选择性RET抑制剂selpercatinib和pralsetinib获得了监管部门的批准。涵盖领域:近10年来发表的万德替尼、卡博赞替尼、赛尔珀卡替尼和普拉塞替尼在MTC中的疗效和安全性数据。自2010年以来,系统MTC治疗的观点发生了重大变化。然而,到目前为止,应该选择哪种药物作为第一线的问题仍然没有解决。没有建议使用什么作为第二或其他治疗线。肿瘤负荷、进展斜率和MTC症状对治疗决策的影响并没有明确的定义。另一个重要的问题是治疗时间。解决这些问题的循证医学研究,是我们近期的任务。治疗耐受性及其对生活质量的影响,特别是对于非选择性MKIs,也是一个重要问题。
{"title":"Recent advances in precision medicine for the treatment of medullary thyroid cancer","authors":"J. Krajewska, A. Kukulska, M. Oczko-Wojciechowska, B. Jarzab","doi":"10.1080/23808993.2021.1964952","DOIUrl":"https://doi.org/10.1080/23808993.2021.1964952","url":null,"abstract":"ABSTRACT Introduction The discovery of a pivotal role of tyrosine kinases in the pathogenesis of medullary thyroid carcinoma (MTC) opened up new options in the systemic treatment of advanced disease. During the last decade, two multikinase inhibitors (MKIs) – vandetanib and cabozantinib and, more recently, two potent, selective RET inhibitors selpercatinib and pralsetinib gained regulatory approval. Areas covered The data on efficacy and safety of vandetanib, cabozantinib, selpercatinib, and pralsetinib in MTC published during the recent 10 years. Expert opinion The perspectives of systemic MTC treatment have substantially changed since 2010. However, to date, the question of which drug should be chosen as the first line remains open. There are no recommendations on what to use as the second or other treatment lines. The impact of tumor burden, progression slope, and the presence of MTC symptoms on treatment-decision making is not unequivocally defined. Another important problem is the treatment duration. EBM (evidence-based medicine) study, resolving these issues, is our task for the nearest future. The treatment tolerability and its impact on the quality of life, particularly regarding nonselective MKIs, is also an essential problem.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"307 - 315"},"PeriodicalIF":1.2,"publicationDate":"2021-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42647580","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
Prediction, diagnosis, prevention and treatment: genetic-led care of patients with diabetes 预测、诊断、预防和治疗:糖尿病患者的遗传主导护理
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-09-03 DOI: 10.1080/23808993.2021.1970526
W. Tangjittipokin, Nutsakol Borrisut, Patcharapong Rujirawan
ABSTRACT Introduction Diabetes mellitus is a serious metabolic disorder that is associated with high morbidity and mortality, and that affects people worldwide – especially in Asia. Genetics plays an important role in disease development, prevention, and therapeutic approach. Precision medicine in diabetes requires a profound understanding of both genetics and the plethora of mechanisms of diabetes. Areas covered This review comprehensively describes and discusses the recent advances in genetics-based prediction, diagnosis, prevention, and treatment of diabetes, and the potential impact of these advances on precision medicine, especially in the Asian population. We analyzed the available literature from the PubMed Central® (PMC) archive. Expert opinion Advanced genetic knowledge can lead to the precise diagnosis, improved risk prediction, diabetes prevention, and genetics-based diabetes treatment. Increased understanding, availability, and affordability of genetics-based strategies will improve the quality of life of those with and those at risk for developing diabetes.
糖尿病是一种严重的代谢性疾病,具有高发病率和高死亡率,影响全球人群,尤其是亚洲人群。遗传学在疾病的发展、预防和治疗方法中起着重要作用。糖尿病的精准医学需要对遗传学和糖尿病的多种机制都有深刻的了解。本综述全面描述和讨论了基于遗传学的糖尿病预测、诊断、预防和治疗的最新进展,以及这些进展对精准医学的潜在影响,特别是对亚洲人群的潜在影响。我们分析了PubMed Central®(PMC)存档中的可用文献。先进的遗传知识可以导致精确的诊断,改进的风险预测,糖尿病预防和基于遗传学的糖尿病治疗。提高对遗传学策略的理解、可获得性和可负担性,将改善糖尿病患者和糖尿病高危人群的生活质量。
{"title":"Prediction, diagnosis, prevention and treatment: genetic-led care of patients with diabetes","authors":"W. Tangjittipokin, Nutsakol Borrisut, Patcharapong Rujirawan","doi":"10.1080/23808993.2021.1970526","DOIUrl":"https://doi.org/10.1080/23808993.2021.1970526","url":null,"abstract":"ABSTRACT Introduction Diabetes mellitus is a serious metabolic disorder that is associated with high morbidity and mortality, and that affects people worldwide – especially in Asia. Genetics plays an important role in disease development, prevention, and therapeutic approach. Precision medicine in diabetes requires a profound understanding of both genetics and the plethora of mechanisms of diabetes. Areas covered This review comprehensively describes and discusses the recent advances in genetics-based prediction, diagnosis, prevention, and treatment of diabetes, and the potential impact of these advances on precision medicine, especially in the Asian population. We analyzed the available literature from the PubMed Central® (PMC) archive. Expert opinion Advanced genetic knowledge can lead to the precise diagnosis, improved risk prediction, diabetes prevention, and genetics-based diabetes treatment. Increased understanding, availability, and affordability of genetics-based strategies will improve the quality of life of those with and those at risk for developing diabetes.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"361 - 374"},"PeriodicalIF":1.2,"publicationDate":"2021-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45211627","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
Brigatinib as a treatment of ALK-positive non-small cell lung cancer 布加替尼治疗alk阳性非小细胞肺癌
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-07-19 DOI: 10.1080/23808993.2021.1954907
E. De Carlo, B. Stanzione, A. Del Conte, A. Revelant, A. Bearz
ABSTRACT Introduction Anaplastic lymphoma kinase (ALK) gene rearrangements and resulting fusion proteins occur in 3%-7% of patients with non-small-cell lung cancer (NSCLC), conferring sensitivity to treatment with ALK Tyrosine-Kinase Inhibitors. After the first-generation ALK inhibitor crizotinib, newer generation ALK inhibitors have been developed and showed greater potency and brain penetration in both crizotinib-naïve and crizotinib-refractory advanced NSCLC patients. Areas covered Brigatinib is a potent and highly selective second generation ALK inhibitor. Brigatinib is approved by Food and Drug Administration and European Medicines Agency for ALK-positive advanced NSCLC patients previously treated with crizotinib and more recently as first-line treatment in naïve patients. In the current review we present an up-to-date overview of the current and evolving clinical data regarding the biology of the disease, the emerging clinical decision-making and the targeted therapy options in advanced NSCLC harboring ALK translocation, especially focusing on the activity of brigatinib. Expert opinion Brigatinib shows a deep systemic and intracranial efficacy in both naïve and refractory metastatic ALK-positive NSCLC patients in the clinical studies, which marked its clinical development. Given its efficacy and tolerability brigatinib could be an excellent therapeutic option for the treatment of advanced ALK-positive NSCLC patients.
间变性淋巴瘤激酶(ALK)基因重排和由此产生的融合蛋白发生在3%-7%的非小细胞肺癌(NSCLC)患者中,赋予ALK酪氨酸激酶抑制剂治疗敏感性。继第一代ALK抑制剂克唑替尼之后,新一代ALK抑制剂已被开发出来,并在crizotinib-naïve和克唑替尼难治性晚期NSCLC患者中显示出更大的效力和脑穿透性。布加替尼是一种有效的、高选择性的第二代ALK抑制剂。Brigatinib已被美国食品和药物管理局(fda)和欧洲药品管理局(ema)批准用于alk阳性晚期NSCLC患者,此前曾接受过克里唑替尼治疗,最近作为naïve患者的一线治疗。在当前的综述中,我们介绍了最新的关于疾病生物学、新出现的临床决策和晚期非小细胞肺癌ALK易位的靶向治疗选择的当前和不断发展的临床数据,特别是关注布加替尼的活性。专家意见布里加替尼在临床研究中对naïve和难治性转移性alk阳性NSCLC患者均显示出深度全身和颅内疗效,标志着其临床发展。鉴于其疗效和耐受性,布加替尼可能是治疗晚期alk阳性NSCLC患者的一种极好的治疗选择。
{"title":"Brigatinib as a treatment of ALK-positive non-small cell lung cancer","authors":"E. De Carlo, B. Stanzione, A. Del Conte, A. Revelant, A. Bearz","doi":"10.1080/23808993.2021.1954907","DOIUrl":"https://doi.org/10.1080/23808993.2021.1954907","url":null,"abstract":"ABSTRACT Introduction Anaplastic lymphoma kinase (ALK) gene rearrangements and resulting fusion proteins occur in 3%-7% of patients with non-small-cell lung cancer (NSCLC), conferring sensitivity to treatment with ALK Tyrosine-Kinase Inhibitors. After the first-generation ALK inhibitor crizotinib, newer generation ALK inhibitors have been developed and showed greater potency and brain penetration in both crizotinib-naïve and crizotinib-refractory advanced NSCLC patients. Areas covered Brigatinib is a potent and highly selective second generation ALK inhibitor. Brigatinib is approved by Food and Drug Administration and European Medicines Agency for ALK-positive advanced NSCLC patients previously treated with crizotinib and more recently as first-line treatment in naïve patients. In the current review we present an up-to-date overview of the current and evolving clinical data regarding the biology of the disease, the emerging clinical decision-making and the targeted therapy options in advanced NSCLC harboring ALK translocation, especially focusing on the activity of brigatinib. Expert opinion Brigatinib shows a deep systemic and intracranial efficacy in both naïve and refractory metastatic ALK-positive NSCLC patients in the clinical studies, which marked its clinical development. Given its efficacy and tolerability brigatinib could be an excellent therapeutic option for the treatment of advanced ALK-positive NSCLC patients.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"299 - 305"},"PeriodicalIF":1.2,"publicationDate":"2021-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2021.1954907","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44886454","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
MSI-H/dMMR and cancer immunotherapy: current state and future implications MSI-H/dMMR与癌症免疫疗法的现状及未来意义
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-06-30 DOI: 10.1080/23808993.2021.1946391
A. Rizzo, A. Ricci, G. Gadaleta-Caldarola
Immunotherapy has revolutionized the treatment landscape of several hematological and solid tumors by producing unprecedented algorithm shifts in a relatively short period of time [1]. However, immune checkpoint inhibitors (ICIs) have been suggested to be effective in approximately onethird of all cancer patients, with the antitumor activity of immunotherapy varying among different malignancies [2]. Thus, the identification of potential responders has recently become one of the key challenges in medical oncology, since there is an urgent need to develop reliable biomarkers that could guide clinicians in patient selection [3,4]. In fact, several predictors of response to ICIs have been tested and evaluated, three of whom have been approved by the United States Food and Drug Administration (FDA): programmed death ligand 1 (PD-L1), tumor mutational burden (TMB), and microsatellite instability/defective mismatch repair (MSI/dMMR) [5]. Notably enough, all these predictors present notable differences in terms of methodology and specificity as well as strengths and weaknesses. Other potentially useful elements as predictors and/or for prognostic stratification are under evaluation, including tumorinfiltrating lymphocytes (TILs) [6]. Two years after the landmark approval of PD-L1 as predictive biomarker in non-small cell lung cancer (NSCLC), pembrolizumab was approved by the FDA for the treatment of patients with MSI-high (MSI-H)/dMMR advanced solid tumors in 2017 [7]. In particular, this approval was based on the results observed in MSI-H/dMMR malignancies across five clinical trials [7]. In fact, in these studies, the PD-1 inhibitor pembrolizumab reported an overall response rate (ORR) and a complete response (CR) rate of 39.6% and 7%, respectively, in MSI-H/dMMR solid tumors; in addition, the 78% of responders presented duration of response of 6 months or longer. Notably enough, the approval of MSI-H had some historical significance, being the first ‘orphan’ approval of a biomarker, regardless of histology and tumor type. From a molecular point of view, dMMR malignancies accumulate mutations across the genome, leading to the formation of neoantigens as well as the activation of antitumor responses [8]. Mismatch errors are particularly frequent in short tandem repeats, and thus, mutations are more commonly observed in microsatellite regions, a condition termed as MSI. Three different testing methods are available for detecting MSI-H/dMMR status: polymerase chain reaction (PCR) and next-generation sequencing (NGS) for MSI-H while dMMR is commonly determined through immunohistochemistry (IHC) [9]. Two PCR panels are more frequently used in clinical practice to determine MSI-H, the first of which is known as the Bathesda panel, including two mononucleotide (BAT-25 and BAT-26) and three dinucleotide (D5S346, D2S123, and D17S250) repeats [10]. Of note, both cancer and paired normal tissue are necessary for the evaluation of MSI-H using this panel. Conversel
免疫疗法在相对较短的时间内产生了前所未有的算法变化,从而彻底改变了几种血液学和实体瘤的治疗格局[1]。然而,免疫检查点抑制剂(ICIs)被认为对大约三分之一的癌症患者有效,免疫疗法的抗肿瘤活性因不同的恶性肿瘤而异[2]。因此,识别潜在反应者最近已成为医学肿瘤学的关键挑战之一,因为迫切需要开发可靠的生物标志物,指导临床医生选择患者[3,4]。事实上,已经对ICIs反应的几种预测因子进行了测试和评估,其中三种已获得美国食品药品监督管理局(FDA)的批准:程序性死亡配体1(PD-L1)、肿瘤突变负荷(TMB)和微卫星不稳定性/缺陷错配修复(MSI/dMMR)[5]。值得注意的是,所有这些预测因素在方法、特异性以及优势和劣势方面都存在显著差异。作为预测因素和/或预后分层的其他潜在有用因素正在评估中,包括肿瘤过滤淋巴细胞(TIL)[6]。在PD-L1作为非小细胞肺癌癌症预测性生物标志物获得里程碑式批准两年后,2017年,美国食品药品监督管理局批准pembrolizumab用于治疗MSI-high(MSI-H)/dMMR晚期实体瘤患者[7]。特别是,这一批准是基于在五项临床试验中观察到的MSI-H/dMMR恶性肿瘤的结果[7]。事实上,在这些研究中,PD-1抑制剂pembrolizumab在MSI-H/dMMR实体瘤中的总有效率(ORR)和完全有效率(CR)分别为39.6%和7%;此外,78%的应答者的应答持续时间为6个月或更长。值得注意的是,MSI-H的批准具有一定的历史意义,是第一个生物标志物的“孤儿”批准,无论组织学和肿瘤类型如何。从分子角度来看,dMMR恶性肿瘤在整个基因组中积累突变,导致新抗原的形成以及抗肿瘤反应的激活[8]。错配错误在短串联重复中特别常见,因此,突变在微卫星区域更常见,这种情况被称为MSI。有三种不同的检测方法可用于检测MSI-H/dMMR状态:MSI-H的聚合酶链式反应(PCR)和下一代测序(NGS),而dMMR通常通过免疫组织化学(IHC)测定[9]。在临床实践中,两个PCR组更频繁地用于测定MSI-H,其中第一个被称为Bathesda组,包括两个单核苷酸(BAT-25和BAT-26)和三个二核苷酸(D5S346、D2S123和D17S250)重复[10]。值得注意的是,癌症和配对的正常组织对于使用该面板评估MSI-H是必要的。相反,第二组是基于对五个多A单核苷酸重复序列(BAT-25、BAT-26、NR-21、NR-24和NR-27)的评估,与前一组相比显示出更高的特异性和敏感性[11]。此外,该面板不需要正常组织,如果五次重复中至少有两次失去稳定性,则恶性肿瘤被确定为MSI-H。然而,目前通过PCR对MSI-H的评估是基于对选定数量的微卫星的分析,因此,在0.5%至10%的病例中观察到假阴性结果[12]。此外,MSI-H的患病率因肿瘤类型而异,在几种恶性肿瘤中没有可用的数据,包括肾细胞癌和黑色素瘤;相反,其他恶性肿瘤的患病率约为1-2%[13]。NGS方法已被评估,以克服与通过PCR评估MSI-H/dMMR相关的限制[14]。事实上,这种基于肿瘤基因组或全外显子组测序的新检测方法有可能评估几种不同类型的微卫星;此外,NGS可用于所有恶性肿瘤,并能够评估TMB。因此,NGS对MSI-H/dMMR的评估也可以整合TMB[15];然而,NGS存在一些缺点,包括成本高和缺乏广泛的可用性。IHC是另一种常用于通过评估MLH1、MSH2、MSH6和PMS2来确定dMMR的方法,其中至少一种MMR蛋白的表达损失被定义为dMMR[16]。IHC的优势包括其简单性和成本;此外,IHC非常可用,并且可以在所有中心轻松完成。然而,IHC有一些重要的弱点,包括
{"title":"MSI-H/dMMR and cancer immunotherapy: current state and future implications","authors":"A. Rizzo, A. Ricci, G. Gadaleta-Caldarola","doi":"10.1080/23808993.2021.1946391","DOIUrl":"https://doi.org/10.1080/23808993.2021.1946391","url":null,"abstract":"Immunotherapy has revolutionized the treatment landscape of several hematological and solid tumors by producing unprecedented algorithm shifts in a relatively short period of time [1]. However, immune checkpoint inhibitors (ICIs) have been suggested to be effective in approximately onethird of all cancer patients, with the antitumor activity of immunotherapy varying among different malignancies [2]. Thus, the identification of potential responders has recently become one of the key challenges in medical oncology, since there is an urgent need to develop reliable biomarkers that could guide clinicians in patient selection [3,4]. In fact, several predictors of response to ICIs have been tested and evaluated, three of whom have been approved by the United States Food and Drug Administration (FDA): programmed death ligand 1 (PD-L1), tumor mutational burden (TMB), and microsatellite instability/defective mismatch repair (MSI/dMMR) [5]. Notably enough, all these predictors present notable differences in terms of methodology and specificity as well as strengths and weaknesses. Other potentially useful elements as predictors and/or for prognostic stratification are under evaluation, including tumorinfiltrating lymphocytes (TILs) [6]. Two years after the landmark approval of PD-L1 as predictive biomarker in non-small cell lung cancer (NSCLC), pembrolizumab was approved by the FDA for the treatment of patients with MSI-high (MSI-H)/dMMR advanced solid tumors in 2017 [7]. In particular, this approval was based on the results observed in MSI-H/dMMR malignancies across five clinical trials [7]. In fact, in these studies, the PD-1 inhibitor pembrolizumab reported an overall response rate (ORR) and a complete response (CR) rate of 39.6% and 7%, respectively, in MSI-H/dMMR solid tumors; in addition, the 78% of responders presented duration of response of 6 months or longer. Notably enough, the approval of MSI-H had some historical significance, being the first ‘orphan’ approval of a biomarker, regardless of histology and tumor type. From a molecular point of view, dMMR malignancies accumulate mutations across the genome, leading to the formation of neoantigens as well as the activation of antitumor responses [8]. Mismatch errors are particularly frequent in short tandem repeats, and thus, mutations are more commonly observed in microsatellite regions, a condition termed as MSI. Three different testing methods are available for detecting MSI-H/dMMR status: polymerase chain reaction (PCR) and next-generation sequencing (NGS) for MSI-H while dMMR is commonly determined through immunohistochemistry (IHC) [9]. Two PCR panels are more frequently used in clinical practice to determine MSI-H, the first of which is known as the Bathesda panel, including two mononucleotide (BAT-25 and BAT-26) and three dinucleotide (D5S346, D2S123, and D17S250) repeats [10]. Of note, both cancer and paired normal tissue are necessary for the evaluation of MSI-H using this panel. Conversel","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"345 - 347"},"PeriodicalIF":1.2,"publicationDate":"2021-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2021.1946391","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49328026","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
An evaluation of selumetinib for the treatment of neurofibromatosis type 1-associated symptomatic, inoperable plexiform neurofibromas 评价selumetinib治疗1型神经纤维瘤病相关症状,不能手术的丛状神经纤维瘤
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-05-31 DOI: 10.1080/23808993.2021.1917989
L. Metrock, M. Lobbous, B. Korf
ABSTRACT Introduction: Plexiform neurofibromas (PNs) are present in up to half of patients with neurofibromatosis type 1 (NF1). PNs consist of a proliferation of abnormal cells in the nerve sheath and can lead to significant comorbidities. Most PNs are diagnosed in early childhood when the most rapid growth rate generally occurs. Historically there has been a paucity of effective treatment options for patients with PNs, with surgery being the standard of care. As knowledge has increased regarding the NF1 gene and the function of its protein product neurofibromin, therapies targeting the Ras signaling pathway have been tested, first in preclinical models and subsequently in clinical trials. Areas covered: This review focuses on selumetinib (KOSELUGOTM; AZD6244, ARRY-142,886) and the management of PNs. A literature search was undertaken using PubMed with keywords ‘neurofibromatosis,’ ‘plexiform neurofibromas,’ ‘selumetinib,’ and ‘MEK inhibitor.’ Expert opinion: Selumetinib is the first FDA approved drug for the treatment of PNs. Prior to its development, options for patients with PNs causing morbidity were limited. Surgical intervention can be difficult, debilitating, and often futile. With the efficacy seen in the phase 1 and 2 trials for patients with NF1-associated PNs, the outlook has become one of the hope and excitement.
摘要简介:丛状神经纤维瘤(PNs)存在于多达一半的1型神经纤维瘤病(NF1)患者中。PNs由神经鞘中异常细胞的增殖组成,可导致显著的合并症。大多数PNs是在儿童早期被诊断出来的,此时生长速度通常最快。从历史上看,PNs患者缺乏有效的治疗选择,手术是护理的标准。随着对NF1基因及其蛋白质产物神经纤维蛋白功能的了解不断增加,针对Ras信号通路的疗法已经进行了测试,首先是在临床前模型中,然后是在临床试验中。涵盖的领域:本综述侧重于selumetinib(KOSELUGOTM;AZD6244,ARRY-142886)和PN的管理。使用PubMed进行文献检索,关键词为“神经纤维瘤病”、“丛状神经纤维瘤”、“selumetinib”和“MEK抑制剂”专家意见:赛卢美替尼是美国食品药品监督管理局批准的第一种治疗PNs的药物。在其开发之前,PNs导致发病的患者的选择是有限的。手术干预可能很困难,使人衰弱,而且往往是徒劳的。随着NF1相关PNs患者的1期和2期试验的疗效,前景已成为希望和兴奋之一。
{"title":"An evaluation of selumetinib for the treatment of neurofibromatosis type 1-associated symptomatic, inoperable plexiform neurofibromas","authors":"L. Metrock, M. Lobbous, B. Korf","doi":"10.1080/23808993.2021.1917989","DOIUrl":"https://doi.org/10.1080/23808993.2021.1917989","url":null,"abstract":"ABSTRACT Introduction: Plexiform neurofibromas (PNs) are present in up to half of patients with neurofibromatosis type 1 (NF1). PNs consist of a proliferation of abnormal cells in the nerve sheath and can lead to significant comorbidities. Most PNs are diagnosed in early childhood when the most rapid growth rate generally occurs. Historically there has been a paucity of effective treatment options for patients with PNs, with surgery being the standard of care. As knowledge has increased regarding the NF1 gene and the function of its protein product neurofibromin, therapies targeting the Ras signaling pathway have been tested, first in preclinical models and subsequently in clinical trials. Areas covered: This review focuses on selumetinib (KOSELUGOTM; AZD6244, ARRY-142,886) and the management of PNs. A literature search was undertaken using PubMed with keywords ‘neurofibromatosis,’ ‘plexiform neurofibromas,’ ‘selumetinib,’ and ‘MEK inhibitor.’ Expert opinion: Selumetinib is the first FDA approved drug for the treatment of PNs. Prior to its development, options for patients with PNs causing morbidity were limited. Surgical intervention can be difficult, debilitating, and often futile. With the efficacy seen in the phase 1 and 2 trials for patients with NF1-associated PNs, the outlook has become one of the hope and excitement.","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"239 - 246"},"PeriodicalIF":1.2,"publicationDate":"2021-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2021.1917989","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49165386","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
Value of digital biomarkers in precision medicine: implications in cancer, autoimmune diseases, and COVID-19 数字生物标志物在精准医学中的价值:对癌症、自身免疫性疾病和COVID-19的影响
IF 1.2 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2021-05-13 DOI: 10.1080/23808993.2021.1924055
E. Capobianco, Marcus John Beasley
Based on the patient’s characteristics, precision medicine (PM) aims to optimize the time of administration of the most appropriate medicine with the minimum risk of toxicity. This is a multidimensional problem due to the varied disease course and therapeutic responses of patients. General factors, such as genetics, epigenetics, environment, ethnicity, adherence, lifestyle, and diet, determine these outcomes. In clinical trials, some drugs may be not beneficial or even harmful for a given ethnic or co-morbid group. Partial response is also observed outside trials, as the most commonly used drugs show high efficacy in relatively few patients. Therefore, what we call ‘imprecise medicine’ is the first challenge of PM due to the assumption underlying clinical practice that disease treatment and prevention strategies developed at the population level are expected to be accurate when applied at the individual level. The complexity that drives the variations in patient profiles depends on the heterogeneity of information obtained from large volumes of genetic, serological, biochemical, and diagnostic imaging data. These represent dimensions that need harmonization and integration with lifestyle and environmental factors. The second challenge is with assessing the benefits of the data dimensions, such as diagnostic improvements, earlier interventions, increased drug efficiency, and better-targeted treatments. To accommodate the heterogeneity of the etiologies, clinical symptoms, and treatment responses of patients in clinical practice, a revised clinical approach is recommended [1]. The first step is the development of a machine learning (ML)-assisted risk assessment model (see, for instance [2],) followed by the identification of the robust multimodal data-driven prognostic indicators (see, for instance [3],). These two efforts require new strategies for integrating heterogeneous information from different structured and unstructured data sources (electronic health records (EHRs), administrative databases, bioimaging archives, self-quantified measurements, etc.). Big Data has introduced a new paradigm for population-based studies that comes with challenges. For instance, the validity of such studies is based on the diagnostic accuracy used for all cases. A critical problem is the variability of the methods used to perform validations. Currently, there are challenges with validating most disease classification algorithms, and this complicates the assessment of their potential for population studies. Model validation facilitates safer interpretability of the correlations between diverse data types revealed by the models. Data-centric perspectives of complex diseases facilitate their definition as heterogeneous processes that have multifaceted causes, courses of evolution, treatments, and patient’s disease trajectories from the observed responses to treatment (see [4–6], among many other examples). These trajectories differ with each patient and, therefore
根据患者的特点,精准医学(PM)旨在以最小的毒性风险优化最合适药物的给药时间。由于不同的病程和患者的治疗反应,这是一个多方面的问题。一般因素,如遗传学、表观遗传学、环境、种族、依从性、生活方式和饮食,决定了这些结果。在临床试验中,一些药物可能对特定的种族或合并症群体没有益处,甚至有害。在试验之外也观察到部分反应,因为最常用的药物在相对较少的患者中显示出高效率。因此,我们所说的“不精确医学”是PM的第一个挑战,因为临床实践的基本假设是,在人群水平上制定的疾病治疗和预防策略在应用于个人水平时预计是准确的。驱动患者概况变化的复杂性取决于从大量遗传、血清学、生化和诊断成像数据中获得的信息的异质性。这些代表了需要与生活方式和环境因素协调和整合的维度。第二个挑战是评估数据维度的益处,例如诊断改进、早期干预、提高药物效率和更有针对性的治疗。为了适应临床实践中患者的病因、临床症状和治疗反应的异质性,建议采用一种修订的临床方法。第一步是开发机器学习(ML)辅助的风险评估模型(参见,例如[2]),然后确定稳健的多模态数据驱动的预后指标(参见,例如[3],)。这两项工作需要新的策略来整合来自不同结构化和非结构化数据源(电子健康记录(EHRs)、管理数据库、生物成像档案、自量化测量等)的异构信息。大数据为基于人口的研究引入了一种新的范式,这种范式也带来了挑战。例如,这些研究的有效性是基于对所有病例的诊断准确性。一个关键的问题是用于执行验证的方法的可变性。目前,在验证大多数疾病分类算法方面存在挑战,这使得评估它们在人口研究中的潜力变得复杂。模型验证有助于更安全地解释模型所揭示的不同数据类型之间的相关性。以数据为中心的复杂疾病视角有助于将其定义为异质性过程,这些过程具有多方面的原因、演变过程、治疗方法以及观察到的患者对治疗反应的疾病轨迹(见[4-6]以及许多其他例子)。这些轨迹因患者而异,因此需要精确的方法。我们强调早期干预的必要性,当分子原因/模式仍然可以确定。早期治疗可能会大大降低疾病进展的风险,延长健康。因此,开发更具包容性的数字生物标志物(db)至关重要[7,8],这些数字生物标志物可能反映临床和分子数据的协同作用,以便在干预措施具有最佳成功机会的早期阶段识别疾病并预防未来的损害。DB值应与疾病过程中缩短轨迹长度的能力成正比,这将减少任何疾病触发和临床干预之间的时间窗口,然后发生不可逆转的损害。
{"title":"Value of digital biomarkers in precision medicine: implications in cancer, autoimmune diseases, and COVID-19","authors":"E. Capobianco, Marcus John Beasley","doi":"10.1080/23808993.2021.1924055","DOIUrl":"https://doi.org/10.1080/23808993.2021.1924055","url":null,"abstract":"Based on the patient’s characteristics, precision medicine (PM) aims to optimize the time of administration of the most appropriate medicine with the minimum risk of toxicity. This is a multidimensional problem due to the varied disease course and therapeutic responses of patients. General factors, such as genetics, epigenetics, environment, ethnicity, adherence, lifestyle, and diet, determine these outcomes. In clinical trials, some drugs may be not beneficial or even harmful for a given ethnic or co-morbid group. Partial response is also observed outside trials, as the most commonly used drugs show high efficacy in relatively few patients. Therefore, what we call ‘imprecise medicine’ is the first challenge of PM due to the assumption underlying clinical practice that disease treatment and prevention strategies developed at the population level are expected to be accurate when applied at the individual level. The complexity that drives the variations in patient profiles depends on the heterogeneity of information obtained from large volumes of genetic, serological, biochemical, and diagnostic imaging data. These represent dimensions that need harmonization and integration with lifestyle and environmental factors. The second challenge is with assessing the benefits of the data dimensions, such as diagnostic improvements, earlier interventions, increased drug efficiency, and better-targeted treatments. To accommodate the heterogeneity of the etiologies, clinical symptoms, and treatment responses of patients in clinical practice, a revised clinical approach is recommended [1]. The first step is the development of a machine learning (ML)-assisted risk assessment model (see, for instance [2],) followed by the identification of the robust multimodal data-driven prognostic indicators (see, for instance [3],). These two efforts require new strategies for integrating heterogeneous information from different structured and unstructured data sources (electronic health records (EHRs), administrative databases, bioimaging archives, self-quantified measurements, etc.). Big Data has introduced a new paradigm for population-based studies that comes with challenges. For instance, the validity of such studies is based on the diagnostic accuracy used for all cases. A critical problem is the variability of the methods used to perform validations. Currently, there are challenges with validating most disease classification algorithms, and this complicates the assessment of their potential for population studies. Model validation facilitates safer interpretability of the correlations between diverse data types revealed by the models. Data-centric perspectives of complex diseases facilitate their definition as heterogeneous processes that have multifaceted causes, courses of evolution, treatments, and patient’s disease trajectories from the observed responses to treatment (see [4–6], among many other examples). These trajectories differ with each patient and, therefore","PeriodicalId":12124,"journal":{"name":"Expert Review of Precision Medicine and Drug Development","volume":"6 1","pages":"235 - 238"},"PeriodicalIF":1.2,"publicationDate":"2021-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1080/23808993.2021.1924055","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43378046","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
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