Issue highlights—September 2023

IF 2.3 3区 医学 Q3 MEDICAL LABORATORY TECHNOLOGY
Professor Frederic I. Preffer
{"title":"Issue highlights—September 2023","authors":"Professor Frederic I. Preffer","doi":"10.1002/cyto.b.22145","DOIUrl":null,"url":null,"abstract":"<p>[Color figure can be viewed at wileyonlinelibrary.com]</p><p>Despite the reality that my entire career has been spent devoted to flow cytometry, it is undeniable to me that the gold standard remains an image of the cell, and when visualized within tissue, the architectural relationships between cells. Thus my keen interest in bringing the Amnis technology into my research laboratory years ago and my current enthusiasm for the cell-imaging capacities of more recent technologies such as those from Becton-Dickinson (Cell-View), ThermoFisher (Attune CytPix) and other imaging flow cytometers currently or soon available. In this issue I am therefore extremely interested in Cyclic Analysis of Single-Cell Subsets and Tissue Territories (CASSATT) which identifies scanned slide images through multiple staining rounds, segments nuclei and assesses marker expression on each detected cell (Brockman et al., <span>2023</span>). Cyclic immunochemistry (cycIHC) exploits multiple rounds of immunostaining and imaging for mapping and locating cells of interest with the speed and simplicity of brightfield microscopy, making the collection of entire tissue sections and slides possible. CASSATT employs registered scanned slide images across all rounds of staining, segments individual nuclei, and measures marker expression on each detected cell. It further explores the spatial relationships between cell populations and the odds of interaction frequencies between cell populations within tissue regions, helping to identify cells that interact or do not interact. In this report, a test dataset of six glioblastoma tissue sections were cyclically stained for eight biomarkers for a total of 48 scanned slide images. The authors describe an efficient workflow that provided answers to questions commonly encountered in discovery research in tissue specimens, such as the overall abundance of cells of interest within a tissue section as well as whether groups of cells were in close proximity. CASSATT gathered together and streamlined all steps necessary to produce single cell expression information from cycIHC datasets and did so utilizing an open source environment. In addition, CASSATT systematically analyzed the spatial relationships between cell populations and via unsupervised algorithms, identified clusters of cell niches.</p><p>The persistence of measurable residual disease (MRD) is a strong indicator for adverse outcomes in acute myeloid leukemia (AML) and has been shown to be a valid surrogate marker for disease-free survival and overall survival, irrespective of patients' age, AML subtype, sample type, time of MRD assessment and MRD detection method (Short et al., <span>2020</span>). Other hematopoietic malignancies such as B- and plasma cell lineages also benefit from MRD diagnostic assays (Chen et al., <span>2023</span>; Gao et al., <span>2023</span>; McMillan et al., <span>2023</span>; Zhou et al., <span>2023</span>). In a study by Wang et al. (<span>2023</span>) their experience working under CLSI HL62 guidelines and FDA IDE (investigational device exemption) approval in the validation of a 12-color AML MRD flow cytometry MRD assay is shared, including the details of panel design, analysis and interpretation.</p><p>Prior to their recent cytometer upgrade to 12-marker capacity permitting more expansive testing, these investigators utilized an eight-marker AML MRD test designed to discriminate between normal and abnormal myelomonocytic precursors using analytical approaches combining the detection of leukemia-associated immunophenotypes (LAIP) and/or the identification of deviation from normal (DfN) methods. The eight-color assay was cross correlated with molecular testing, clinical outcomes, and resulted in multiple publications (Ouyang et al., <span>2015</span>, <span>2016</span>; Xu et al., <span>2017</span>).</p><p>In the current study, the assay accuracy was assessed by testing known positive and negative samples and correlating with the results of molecular genetic testing and follow-up bone marrow examination. The limit of detection (LOD) and limit of quantification (LOQ) were validated to a level between 0.01% and 0.1%, dependent on the numbers of cells evaluated and the degree of deviation from normal phenotypes. Assay linearity, precision and carry over studies were found to be acceptable. The clinical validity of the assay was tested in 61 patients in order that “trueness” could be determined by correlating with concurrent molecular genetic testing and or follow-up bone marrow examination results; the clinical test concordance was 93%, specificity 98% and sensitivity 83%. Ultimately, the most challenging aspects of the assay involved discerning differences between pre-leukemic cells (persistent clonal hematopoiesis) or underlying myelodysplastic clones from AML MRD with immunophenotypic switch or subclone selection, highlighting the need for further characterization of abnormal blasts bearing the potential for relapse.</p><p>Processing as many monoclonal antibodies as possible in one staining tube in the clinical flow cytometry laboratory has been an on-going aspiration over my entire professional career. It is not as much of the “flow-geek” in me that wants such capacity but rather my contention that such practice clearly provides better medical care to our patients. This is especially true in the diagnosis of hematologic malignancies, where I contend that the tumors “…do not read the same books and journals” that we do related to linage markers, but rather do whatever they can to obfuscate their lineages and confuse us with their “unfaithful” antigenic co-expressions. The more markers we can apply to such malignancies, along with built in internal positive and negative controls (provided by comingling normal cells), the better diagnosticians we become. These are not the only advantages of minimizing the number of staining tubes, in that when samples are pauci-cellular, more diagnostic information also becomes further available utilizing the least number of tubes or wells cells are processed in. The more analytic power the clinical flow cytometry laboratory can harness, the more flexibility we can offer and better questions we can ask such as those described just above related to MRD as well as so many other areas (Estevam et al., <span>2021</span>; Quirós-Caso et al., <span>2022</span>; Shameli &amp; Roshan, <span>2022</span>; Sanjabi &amp; Lear, <span>2021</span>). Such hardware, reagents and software already exists in our research flow cytometry laboratories; we are all left waiting for these to move fully into the clinical realm, and to see advances in this area brings us all closer to far better laboratory practice.</p><p>In the submission of Hammerich et al. (<span>2023</span>), we learn of the application of a three-laser spectral flow cytometer demonstrating the analysis of a 31-marker clinically oriented testing panel. The standard 3-laser Aurora utilized by this group resolved 31 fluorochromes excited off a 405, 488, and 640 nm lasers. All fluorochromes and titrated antibodies used in the study were commercially available simplifying modification of the panel and facilitating replacement of markers of interest with others that might be more appropriate. In fact since not all detectors were utilized in this study, it is possible to hypothesize that in the future a few more conjugated antibodies might possibly expand the breadth of this platform's analytic capacity. In conclusion, I find this to be a great moment in clinical flow diagnostics… except of course that I want at least one if not two- to -three more lasers in the instrument than those discussed in this article .</p><p>It is my privilege and honor to usher into our Journal a new type of peer-reviewed submission we refer to as the “Best Practice” category. While presently new to our Journal pages, in past years such informative pieces in the form of numbered Modules have been available to the clinical flow laboratory community, originating from our own ICCS Quality and Standards Committee. The current submission from Devitt et al. (<span>2023</span>) underscores the importance of flow cytometric assay validation which provides confidence that such assays yield reliable results that can be trusted by clinical caregivers in their determining critical medical decisions.</p><p>For example, the authors provide our readership much needed descriptions, explanations and distinguishing characteristics of both IVD (in vitro diagnostic) or LDT (laboratory developed) tests. IVD tests are developed by manufacturers who optimize, validate, and submit such assays to a regulatory body, such as the FDA. The regulatory body approves the validation and clears it for clinical use permitting the manufacturer to sell the assay to laboratories for testing patient samples. Laboratories must follow the manufacturer's described standard operating procedures and verify that they can reproduce the manufacturer's performance specifications in their lab.</p><p>In contrast to IVD tests, Devitt et al. (<span>2023</span>) focusses on LDT assays which are developed, optimized and validated in individual laboratories using that specific laboratories equipment, reagents and staff. There is currently no requirement for an outside regulatory body to approve LDTs in the United States, although federal legislation to change this paradigm (VALID act) has been suggested and such potential changes must be closely monitored by many impacted medical societies. The laboratory must validate performance specifications unique to their in-house-developed assay. Once validated, the laboratory can perform the assay on patient samples; however, tests may not be sold to another facility. Any change to an IVD assay that deviates from the manufacturer instructions may render the test an LDT and require validation. The authors continue with other aspects of LDTs such as describing their reporting structures (quantitative, semi- quantitative, and qualitative or mixed) and providing examples of each. The concepts of testing accuracy, precision, limits of detectability, stability along with helpful examples of each concept round out this incredibly helpful reference document. I trust this is a great start to our Journal publishing's future contributions in this new category!</p><p>Four “Letters to the Editor” generally related to hematologic malignancy round off this issue of Clinical Cytometry (Li et al., <span>2023</span>; Martin-Moro &amp; Garcia-Vela, <span>2023</span>; Panda et al., <span>2023a</span>, <span>2023b</span>).</p><p>With that I would like to thank all who submit articles to our Journal, as well as our splendid Associate Editors and talented Editorial Board who are involved in the important task of reviewing submitted articles. Additionally, hats off to our friends at Wiley, the ICCS and ESCCA. And a special thanks to Doris Regal with whom I have the pleasure of working with in making this Journal a success!</p>","PeriodicalId":10883,"journal":{"name":"Cytometry Part B: Clinical Cytometry","volume":null,"pages":null},"PeriodicalIF":2.3000,"publicationDate":"2023-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cyto.b.22145","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cytometry Part B: Clinical Cytometry","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cyto.b.22145","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"MEDICAL LABORATORY TECHNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

[Color figure can be viewed at wileyonlinelibrary.com]

Despite the reality that my entire career has been spent devoted to flow cytometry, it is undeniable to me that the gold standard remains an image of the cell, and when visualized within tissue, the architectural relationships between cells. Thus my keen interest in bringing the Amnis technology into my research laboratory years ago and my current enthusiasm for the cell-imaging capacities of more recent technologies such as those from Becton-Dickinson (Cell-View), ThermoFisher (Attune CytPix) and other imaging flow cytometers currently or soon available. In this issue I am therefore extremely interested in Cyclic Analysis of Single-Cell Subsets and Tissue Territories (CASSATT) which identifies scanned slide images through multiple staining rounds, segments nuclei and assesses marker expression on each detected cell (Brockman et al., 2023). Cyclic immunochemistry (cycIHC) exploits multiple rounds of immunostaining and imaging for mapping and locating cells of interest with the speed and simplicity of brightfield microscopy, making the collection of entire tissue sections and slides possible. CASSATT employs registered scanned slide images across all rounds of staining, segments individual nuclei, and measures marker expression on each detected cell. It further explores the spatial relationships between cell populations and the odds of interaction frequencies between cell populations within tissue regions, helping to identify cells that interact or do not interact. In this report, a test dataset of six glioblastoma tissue sections were cyclically stained for eight biomarkers for a total of 48 scanned slide images. The authors describe an efficient workflow that provided answers to questions commonly encountered in discovery research in tissue specimens, such as the overall abundance of cells of interest within a tissue section as well as whether groups of cells were in close proximity. CASSATT gathered together and streamlined all steps necessary to produce single cell expression information from cycIHC datasets and did so utilizing an open source environment. In addition, CASSATT systematically analyzed the spatial relationships between cell populations and via unsupervised algorithms, identified clusters of cell niches.

The persistence of measurable residual disease (MRD) is a strong indicator for adverse outcomes in acute myeloid leukemia (AML) and has been shown to be a valid surrogate marker for disease-free survival and overall survival, irrespective of patients' age, AML subtype, sample type, time of MRD assessment and MRD detection method (Short et al., 2020). Other hematopoietic malignancies such as B- and plasma cell lineages also benefit from MRD diagnostic assays (Chen et al., 2023; Gao et al., 2023; McMillan et al., 2023; Zhou et al., 2023). In a study by Wang et al. (2023) their experience working under CLSI HL62 guidelines and FDA IDE (investigational device exemption) approval in the validation of a 12-color AML MRD flow cytometry MRD assay is shared, including the details of panel design, analysis and interpretation.

Prior to their recent cytometer upgrade to 12-marker capacity permitting more expansive testing, these investigators utilized an eight-marker AML MRD test designed to discriminate between normal and abnormal myelomonocytic precursors using analytical approaches combining the detection of leukemia-associated immunophenotypes (LAIP) and/or the identification of deviation from normal (DfN) methods. The eight-color assay was cross correlated with molecular testing, clinical outcomes, and resulted in multiple publications (Ouyang et al., 2015, 2016; Xu et al., 2017).

In the current study, the assay accuracy was assessed by testing known positive and negative samples and correlating with the results of molecular genetic testing and follow-up bone marrow examination. The limit of detection (LOD) and limit of quantification (LOQ) were validated to a level between 0.01% and 0.1%, dependent on the numbers of cells evaluated and the degree of deviation from normal phenotypes. Assay linearity, precision and carry over studies were found to be acceptable. The clinical validity of the assay was tested in 61 patients in order that “trueness” could be determined by correlating with concurrent molecular genetic testing and or follow-up bone marrow examination results; the clinical test concordance was 93%, specificity 98% and sensitivity 83%. Ultimately, the most challenging aspects of the assay involved discerning differences between pre-leukemic cells (persistent clonal hematopoiesis) or underlying myelodysplastic clones from AML MRD with immunophenotypic switch or subclone selection, highlighting the need for further characterization of abnormal blasts bearing the potential for relapse.

Processing as many monoclonal antibodies as possible in one staining tube in the clinical flow cytometry laboratory has been an on-going aspiration over my entire professional career. It is not as much of the “flow-geek” in me that wants such capacity but rather my contention that such practice clearly provides better medical care to our patients. This is especially true in the diagnosis of hematologic malignancies, where I contend that the tumors “…do not read the same books and journals” that we do related to linage markers, but rather do whatever they can to obfuscate their lineages and confuse us with their “unfaithful” antigenic co-expressions. The more markers we can apply to such malignancies, along with built in internal positive and negative controls (provided by comingling normal cells), the better diagnosticians we become. These are not the only advantages of minimizing the number of staining tubes, in that when samples are pauci-cellular, more diagnostic information also becomes further available utilizing the least number of tubes or wells cells are processed in. The more analytic power the clinical flow cytometry laboratory can harness, the more flexibility we can offer and better questions we can ask such as those described just above related to MRD as well as so many other areas (Estevam et al., 2021; Quirós-Caso et al., 2022; Shameli & Roshan, 2022; Sanjabi & Lear, 2021). Such hardware, reagents and software already exists in our research flow cytometry laboratories; we are all left waiting for these to move fully into the clinical realm, and to see advances in this area brings us all closer to far better laboratory practice.

In the submission of Hammerich et al. (2023), we learn of the application of a three-laser spectral flow cytometer demonstrating the analysis of a 31-marker clinically oriented testing panel. The standard 3-laser Aurora utilized by this group resolved 31 fluorochromes excited off a 405, 488, and 640 nm lasers. All fluorochromes and titrated antibodies used in the study were commercially available simplifying modification of the panel and facilitating replacement of markers of interest with others that might be more appropriate. In fact since not all detectors were utilized in this study, it is possible to hypothesize that in the future a few more conjugated antibodies might possibly expand the breadth of this platform's analytic capacity. In conclusion, I find this to be a great moment in clinical flow diagnostics… except of course that I want at least one if not two- to -three more lasers in the instrument than those discussed in this article .

It is my privilege and honor to usher into our Journal a new type of peer-reviewed submission we refer to as the “Best Practice” category. While presently new to our Journal pages, in past years such informative pieces in the form of numbered Modules have been available to the clinical flow laboratory community, originating from our own ICCS Quality and Standards Committee. The current submission from Devitt et al. (2023) underscores the importance of flow cytometric assay validation which provides confidence that such assays yield reliable results that can be trusted by clinical caregivers in their determining critical medical decisions.

For example, the authors provide our readership much needed descriptions, explanations and distinguishing characteristics of both IVD (in vitro diagnostic) or LDT (laboratory developed) tests. IVD tests are developed by manufacturers who optimize, validate, and submit such assays to a regulatory body, such as the FDA. The regulatory body approves the validation and clears it for clinical use permitting the manufacturer to sell the assay to laboratories for testing patient samples. Laboratories must follow the manufacturer's described standard operating procedures and verify that they can reproduce the manufacturer's performance specifications in their lab.

In contrast to IVD tests, Devitt et al. (2023) focusses on LDT assays which are developed, optimized and validated in individual laboratories using that specific laboratories equipment, reagents and staff. There is currently no requirement for an outside regulatory body to approve LDTs in the United States, although federal legislation to change this paradigm (VALID act) has been suggested and such potential changes must be closely monitored by many impacted medical societies. The laboratory must validate performance specifications unique to their in-house-developed assay. Once validated, the laboratory can perform the assay on patient samples; however, tests may not be sold to another facility. Any change to an IVD assay that deviates from the manufacturer instructions may render the test an LDT and require validation. The authors continue with other aspects of LDTs such as describing their reporting structures (quantitative, semi- quantitative, and qualitative or mixed) and providing examples of each. The concepts of testing accuracy, precision, limits of detectability, stability along with helpful examples of each concept round out this incredibly helpful reference document. I trust this is a great start to our Journal publishing's future contributions in this new category!

Four “Letters to the Editor” generally related to hematologic malignancy round off this issue of Clinical Cytometry (Li et al., 2023; Martin-Moro & Garcia-Vela, 2023; Panda et al., 2023a, 2023b).

With that I would like to thank all who submit articles to our Journal, as well as our splendid Associate Editors and talented Editorial Board who are involved in the important task of reviewing submitted articles. Additionally, hats off to our friends at Wiley, the ICCS and ESCCA. And a special thanks to Doris Regal with whom I have the pleasure of working with in making this Journal a success!

Abstract Image

2023年9月发布。
尽管我的整个职业生涯都致力于流式细胞术,但对我来说不可否认的是,黄金标准仍然是细胞的图像,当在组织中可视化时,细胞之间的结构关系。因此,多年前我对将Amnis技术引入我的研究实验室产生了浓厚的兴趣,并且我目前对最近技术的细胞成像能力充满热情,例如Becton-Dickinson (Cell-View), ThermoFisher (tune CytPix)和其他目前或即将上市的成像流式细胞仪。因此,在这一期中,我对单细胞亚群和组织区域的循环分析(CASSATT)非常感兴趣,该分析通过多个染色轮、细胞核片段识别扫描的幻灯片图像,并评估每个检测细胞上的标记表达(Brockman等人,2023)。循环免疫化学(cycIHC)利用多轮免疫染色和成像来定位和定位感兴趣的细胞,具有明场显微镜的速度和简单性,使整个组织切片和载玻片的收集成为可能。CASSATT采用在所有染色轮中注册的扫描幻灯片图像,分割单个细胞核,并测量每个检测细胞上的标记表达。它进一步探讨了细胞群之间的空间关系和组织区域内细胞群之间相互作用频率的几率,有助于识别相互作用或不相互作用的细胞。在本报告中,对6个胶质母细胞瘤组织切片的测试数据集进行了8种生物标志物的循环染色,共扫描了48张幻灯片图像。作者描述了一个有效的工作流程,为组织标本发现研究中经常遇到的问题提供答案,例如组织切片中感兴趣的细胞的总体丰度以及细胞群是否接近。CASSATT收集并简化了从cycIHC数据集中产生单细胞表达信息所需的所有步骤,并利用开源环境完成了这一过程。此外,CASSATT系统地分析了细胞群体之间的空间关系,并通过无监督算法识别了细胞生态位集群。可测量残余疾病(MRD)的持续存在是急性髓性白血病(AML)不良结局的一个强有力的指标,已被证明是无病生存和总生存的有效替代标志物,与患者的年龄、AML亚型、样本类型、MRD评估时间和MRD检测方法无关(Short et al., 2020)。其他造血恶性肿瘤,如B细胞和浆细胞谱系也受益于MRD诊断分析(Chen等人,2023;Gao et al., 2023;McMillan et al., 2023;Zhou et al., 2023)。Wang等人(2023)在一项研究中分享了他们在CLSI HL62指南和FDA IDE(研究设备豁免)批准下验证12色AML MRD流式细胞术MRD检测的经验,包括面板设计,分析和解释的细节。在他们最近的细胞仪升级到12个标记容量允许更广泛的测试之前,这些研究人员使用了一个8个标记的AML MRD测试,设计用于区分正常和异常的骨髓单核细胞前体,使用结合检测白血病相关免疫表型(LAIP)和/或识别偏离正常(DfN)方法的分析方法。八色法与分子检测、临床结果交叉相关,并发表了多篇论文(Ouyang et al., 2015,2016;Xu et al., 2017)。在目前的研究中,通过检测已知的阳性和阴性样本,并与分子基因检测和后续骨髓检查的结果相关联,来评估检测的准确性。检测限(LOD)和定量限(LOQ)被验证为0.01%和0.1%之间的水平,取决于评估的细胞数量和与正常表型的偏差程度。检测线性度、精密度和结转研究均可接受。在61例患者中测试了该方法的临床有效性,以便通过与并发分子基因检测和/或随访骨髓检查结果的相关性来确定“真实性”;临床试验一致性93%,特异性98%,敏感性83%。最终,该试验最具挑战性的方面涉及识别白血病前细胞(持续克隆造血)或潜在骨髓增生异常克隆之间的差异,这些克隆来自AML MRD,具有免疫表型开关或亚克隆选择,强调需要进一步表征具有复发可能性的异常母细胞。 在临床流式细胞术实验室的一个染色管中处理尽可能多的单克隆抗体一直是我整个职业生涯的夙愿。与其说我是“心流极客”,不如说是我想要这种能力,而是我认为,这种做法显然为我们的病人提供了更好的医疗服务。在血液学恶性肿瘤的诊断中尤其如此,我认为肿瘤“不阅读与我们所阅读的谱系标记相同的书籍和期刊”,而是尽其所能地混淆它们的谱系,让我们对它们“不忠实的”抗原共表达感到困惑。我们对这类恶性肿瘤应用的标志物越多,以及内置的阳性和阴性对照(由混合的正常细胞提供),我们就越能成为更好的诊断专家。这些并不是减少染色管数量的唯一优点,因为当样品是少细胞的时候,利用最少数量的管或孔处理细胞,更多的诊断信息也变得进一步可用。临床流式细胞术实验室可以利用的分析能力越强,我们可以提供的灵活性就越大,我们可以提出的问题也就越好,比如上面描述的与MRD以及许多其他领域相关的问题(Estevam等人,2021;Quirós-Caso等,2022;Shameli,罗山,2022;Sanjabi,李尔王,2021)。这些硬件、试剂和软件在我们的研究流式细胞仪实验室已经存在;我们都在等待这些技术完全进入临床领域,并看到这一领域的进步使我们更接近更好的实验室实践。在Hammerich等人(2023)提交的报告中,我们了解到三激光光谱流式细胞仪的应用,展示了31个标志物临床导向检测面板的分析。该小组使用的标准3激光极光分辨了405、488和640纳米激光激发的31个荧光色。研究中使用的所有荧光染料和滴定抗体都是市售的,简化了对面板的修改,并便于用其他可能更合适的标记替换感兴趣的标记。事实上,由于本研究中没有使用所有的检测器,因此可以假设,在未来,更多的偶联抗体可能会扩大该平台的分析能力。总之,我发现这是临床流诊断的一个伟大时刻,当然,除了我希望仪器中至少有一个,如果不是两到三个激光器,而不是在这篇文章中讨论的,这是我的荣幸和荣幸,为我们的杂志引入一种新的同行评审的提交,我们称之为“最佳实践”类别。虽然目前我们的期刊页面是新的,但在过去的几年里,这些以编号模块形式提供的信息片段已经提供给临床流动实验室社区,源于我们自己的ICCS质量和标准委员会。Devitt等人(2023)目前提交的报告强调了流式细胞术检测验证的重要性,它为临床护理人员在确定关键医疗决策时产生可靠的结果提供了信心。例如,作者为我们的读者提供了IVD(体外诊断)或LDT(实验室开发)测试急需的描述、解释和区别特征。IVD测试是由制造商开发的,他们优化、验证并将此类检测提交给监管机构,如FDA。监管机构批准验证并批准其临床使用,允许制造商将该分析方法出售给实验室用于检测患者样本。实验室必须遵循制造商描述的标准操作程序,并验证他们可以在实验室中复制制造商的性能规格。与IVD测试相比,Devitt等人(2023)侧重于LDT分析,这些分析是在使用特定实验室设备、试剂和工作人员的单个实验室开发、优化和验证的。在美国,目前不要求外部监管机构批准ldt,尽管已经建议通过联邦立法来改变这种模式(VALID法案),而且许多受影响的医疗协会必须密切监测这种潜在的变化。实验室必须验证其内部开发的分析方法的独特性能规格。一旦验证,实验室就可以对患者样本进行分析;但是,检测不得出售给其他机构。对IVD分析的任何改变,如果偏离了制造商的说明,都可能使该测试成为LDT,需要验证。作者继续介绍ldt的其他方面,例如描述它们的报告结构(定量的、半定量的、定性的或混合的),并提供每种结构的例子。 测试准确性,精度,可检测性限制,稳定性的概念以及每个概念的有用示例使这个非常有用的参考文档更加完善。我相信这是我们期刊出版在这一新领域未来贡献的一个良好开端!《临床细胞术》杂志(Li et al., 2023;Martin-Moro,Garcia-Vela, 2023;Panda et al., 2023a, 2023b)。在此,我要感谢所有向我们杂志投稿的人,以及我们优秀的副编辑和才华横溢的编辑委员会,他们参与了审查投稿文章的重要任务。另外,向我们在Wiley的朋友,ICCS和ESCCA致敬。特别感谢多丽丝·雷加尔,我很高兴与她一起工作,使这本杂志取得了成功!
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
6.80
自引率
32.40%
发文量
51
审稿时长
>12 weeks
期刊介绍: Cytometry Part B: Clinical Cytometry features original research reports, in-depth reviews and special issues that directly relate to and palpably impact clinical flow, mass and image-based cytometry. These may include clinical and translational investigations important in the diagnostic, prognostic and therapeutic management of patients. Thus, we welcome research papers from various disciplines related [but not limited to] hematopathologists, hematologists, immunologists and cell biologists with clinically relevant and innovative studies investigating individual-cell analytics and/or separations. In addition to the types of papers indicated above, we also welcome Letters to the Editor, describing case reports or important medical or technical topics relevant to our readership without the length and depth of a full original report.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信