2024 年 5 月要闻

IF 2.3 3区 医学 Q3 MEDICAL LABORATORY TECHNOLOGY
F. Mandy
{"title":"2024 年 5 月要闻","authors":"F. Mandy","doi":"10.1002/cyto.b.22184","DOIUrl":null,"url":null,"abstract":"<p>Just 50 years ago, in 1974, the first fluorescence-activated cell sorter (FACS) was ready for sale. Becton-Dickenson (BD) with a license from Stanford University introduced the FACS sorting platform, which was called the FACS-1. The Herzenberg group at Stanford patented this new flow cytometry (FC) platform 2 years earlier. To this day the popular acronym “FACS” is misused in that most BD FC are cell analyzers, yet they are all called FACS machines. Whether or not a flow cytometer can sort cells, they all detect receptors bound with fluorescent tags on leukocyte subsets. Herzenberg's brilliant idea to integrate four essential 20th-century discoveries related to cellular metrics into a single platform set the stage for a new phase of complex analytical platforms to support the fight against diseases. They include multi-laser excitation, hybridoma technology for tagging fluorescently tagged monoclonal antibodies, signal processing with fast microchips and multi-channel cell sorting.</p><p>Thanks to rapid access to information, when visiting Paul Robertson's virtual library of Cytometry History at Perdue University, it is possible to appreciate how rapidly flow cytometry has matured in over five decades. In minutes, one learns about the interactions between Mack Fulwyler, Len Herzenberg, Bob Auer, Ceasar Milstein, Howard Shapiro and many other fascinating pioneers of the bio-convergence revolution of the 20th-century. The cell sorting technology uses piezo-based oscillation to charge saline droplet-enveloped cells, which are transported to be analyzed and sorted to isolate leukocyte phenotypes of interest. The droplet formation for cell sorting was Fulwyler's adaptation of technology developed for inkjet printers. With fluorescence activation, cells of interest become visible and available to be sorted for functional verification if required. In the 19th century, philosopher Arthur Schopenhauer said, “Talent hits a target no one can hit; genius hits a target no one else can see.” For a half-century, thanks to Herzenberg's contribution, most of us with talent could see leukocyte subsets with statistical significance. Steady advancements in FC continue, and multi-labeled cells can be analyzed with confidence in clinical FC laboratories with tenacity and some talent. This nostalgic indulgence is now over, and the highlights of this issue are to follow.</p><p>Three original articles and two reports on best practices are covered. The first original article is about a novel optimization method to monitor B-cell maturation antigen-targeted chimeric antigen receptors in peripheral blood. The second is an update in the understanding of the role of CD20<sup>+</sup> T-cells. The third article is about the performance of a novel 8-color panel for measurable residual disease assessment in CLL. The first best practice report validates a T-cell receptor β-chain on the constant region (TRBC) immunophenotyping protocol. This new technology improves the diagnosis of T-cell neoplasm. The other best practice report covers a more comprehensive monocyte subset analysis with flow cytometry, with increased sensitivity.</p><p>“Optimization of a flow cytometry test for routine monitoring of B-cell maturation antigen-targeted CAR in peripheral blood” is about monitoring B-cell maturation with targeted chimeric antigen T-cell receptors. The authors report encouraging news about the overall treatment success with multiple myeloma. Even on occasions when initial treatment fails, resorting to relapsed or refractory disease treatment results are encouraging with the two available FDA-licensed drugs. The authors discuss the need for a biomarker to offer a clinical outcome assessment tool. This is possible because of the availability of two reportable variables: CAR T-cell expansion kinetics with toxicity and the ability to accurately quantify B-cell maturation antigen (BCMA) CAR T-cells from peripheral blood. An earlier report from Das et al. (<span>2022</span>) indicated that immunophenotypic profile situation and post-therapy alteration in antigenic expression are available in normal, reactive, and aberrant plasma cells (NPC, RPC, and APC), respectively, for impact on measurable residual disease (MRD) assessment in multiple myeloma (MM). They concluded that antigenic aberrations on polyclonal PC signify the importance of MRD assay validation on a large cohort under normal and reactive conditions. The Das group focused on a cell-surface protein B-cell maturation antigen: CD269 as it emerged as a promising target for CAR-T cell and MAb therapies in MM. However, the knowledge of the BCMA expression pattern in myeloma patients from the Indian subcontinent was not available at that time. A study presented by Sriram et al. (<span>2022</span>) is an in-depth report of BCMA expression on abnormal plasma cells (APC) in Indian MM patients. In the UK, in a 2022 MRD assessment, MM was not incorporated into routine clinical use outside of trials. A report was released (McMillan et al., <span>2023</span>) about achieving minimal residual disease (MRD) negativity following treatment for multiple myeloma (MM) is associated with improved progression-free and overall survival. They suggest a reference MCF MM MRD method, which was stable for up to 6 days following sample collection, allowing broader access by smaller laboratories to further investigate the predictive value and clinical utility of MRD assessments.</p><p>“The role of CD20<sup>+</sup> T-cells: Insights in human peripheral blood.” This study contributes to the understanding of CD20<sup>+</sup> T-cell function in the proinflammatory cascade. The distribution of CD20<sup>−</sup> T-cells among maturation-associated T-cell compartments (naïve, central memory, transitional memory, effector memory, and effector T-cells), their polarization, activation status, and expression of immune-regulatory proteins were evaluated with flow cytometry. Their functions were assessed by measuring IFN-γ, TNF-α and IL-17 production. Compared with CD20<sup>−</sup> T-cells, CD20<sup>+</sup> T-cells represent a higher proportion of transitional memory cells. CD20<sup>+</sup> T-cells display a proinflammatory phenotype, characterized by the expansion of Th1, Th1/17, and Te1 cell subsets. They have been described and immunophenotypically characterized and purified by cell-sorting from neoplastic cells derived from lymph nodes involving T-cell/histiocyte-rich large B-cell lymphoma (Glynn &amp; Fromm, <span>2020</span>). In 2020, Gatti et al. (<span>2021</span>) published ISCCA's immunophenotypic protocol for monitoring anti-CD20 therapies in autoimmune disorders. Also, in 2020, there was a Letter to the Editor (Gadgeel et al., <span>2020</span>) about CD20<sup>+</sup> T-cells in the mediastinal mass tissue with histologically proven PMLBCL and with flow cytometric analysis. CD20<sup>+</sup> T-cells were identified as CD3, CD7, and CD2 positive cells.</p><p>“A novel 8-color FC panel for measurable residual disease (MRD) to monitor chronic lymphocytic leukemia.” The authors report on quantifiable residual disease (MRD) in chronic lymphocytic leukemia (CLL). This prognostic indicator of CLL is of interest as it has been established as predictive of progression-free survival (PFS) and overall survival (OS) in fixed-duration treatment regimens. The authors did a performance evaluation of their novel 8-color panel against the European Research Initiative on CCL (ERIC) 8-color assay. It performed well when compared with ERIC. Stage 0 hematogenous (Hgs) and their association with CD19+ Hgs in anti-CD19 therapy and conventional chemotherapy were reported (Ramalingam et al., <span>2024</span>). There is the potential to mistake a patient with residual disease if the patient received treatment that included anti-CD19 therapy as it can mimic residual Hgs. Two recent publications are addressing the use of AI to deal with the complexity of 8-color FC assay. AI analysis was used to assess measurable residual disease (Shopsowitz et al., <span>2024</span>). However, Bazinet et al. (<span>2023</span>) treated MRD after acute myeloid leukemia. They evaluated DeepFlow, an AI-assisted FC analysis software, to determine if it is suitable for performing automated clustering and identifying cell populations. They concluded that the AI-assisted analysis generates CLL MRD results similar to expert manual analysis. Their program also creates a discussion about the strengths and limitations of the AI approach.</p><p>“TRBC1 FC assay development validation and reporting considerations.” The authors reassessed the current practice of T-cell clonality by flow cytometry. They explain that because a significant gap existed between surface light chains on B-cells, for a long time it was a dependable marker for workup for hematological samples compared with T-cell oncology workups. T-cell receptor β-chain constant region (TRBC) was introduced as an alternative to TGR-V. Until recently, the assessment of clonal T-cells relied on varying marker expression levels, often providing non-specific interpretation. However, the discovery of TRBC1 puts clonal T-cell assessment by flow cytometry much closer to the clinical evaluation with B-cell clonal analysis. Recently Castillo et al. (<span>2024</span>) published a report about diagnosing T-cell non-Hodgkin lymphomas (NHL) with a MAb specific for T-cell receptor β-constant region1 (TRBC1). A study by Lee et al. (<span>2024</span>) covered an optimized whole-blood assay using human recombinant sBCMA to monitor the presence of BCMA CAR T-cells in peripheral blood. Such monitoring can be incorporated into clinical diagnostics, including the proportions of T-cells expressing the anti-BCMA CAR and absolute counts of CAR T cells/μL blood. Wang et al. (<span>2023</span>) reported a method for optimizing fluorochromes for TRBC1 to improve separation between positive and negative cells.</p><p>“Technical gating and interpretation recommendations for partitioning circulating monocyte subsets assessed by flow cytometry.” In the early days of clinical FC, monocytes were a hindrance as they had fewer CD4 MAb receptors than T-helper cells. Hence, they occasionally did compromise CD4 T-cell reporting. A second marker was added to avoid obtaining unreliable T-helper cell counts for HIV disease immunophenotyping (CD3<sup>+</sup>/CD4<sup>+</sup>). Over the past two decades, monocyte assays have been developed to detect CMML. The partitioning of circulatory monocyte subsets by flow cytometry was recommended by Wagner-Ballon et al. (<span>2023</span>). There are three monocyte subsets: cMo(CD14<sup>++</sup>/CD16<sup>−</sup>), iMo(CD14<sup>++</sup>/CD16<sup>+</sup>), and ncMo(CD14<sup>low/−</sup>/CD16<sup>+</sup>). In 2021, Devitt et al. (<span>2023</span>) disclosed the validation protocol for monocytic leukemia diagnosis at the European LeukemiaNet International MDS-Flow Cytometry Working Group (ELN iMDS-Flow). A year later, Jurado et al. (<span>2023</span>) published an optimization of monocyte gating protocol and summarized an enhanced validation approach using qualitative and semiquantitative flow cytometry technology.</p>","PeriodicalId":10883,"journal":{"name":"Cytometry Part B: Clinical Cytometry","volume":null,"pages":null},"PeriodicalIF":2.3000,"publicationDate":"2024-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cyto.b.22184","citationCount":"0","resultStr":"{\"title\":\"Issue highlights—May 2024\",\"authors\":\"F. Mandy\",\"doi\":\"10.1002/cyto.b.22184\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Just 50 years ago, in 1974, the first fluorescence-activated cell sorter (FACS) was ready for sale. Becton-Dickenson (BD) with a license from Stanford University introduced the FACS sorting platform, which was called the FACS-1. The Herzenberg group at Stanford patented this new flow cytometry (FC) platform 2 years earlier. To this day the popular acronym “FACS” is misused in that most BD FC are cell analyzers, yet they are all called FACS machines. Whether or not a flow cytometer can sort cells, they all detect receptors bound with fluorescent tags on leukocyte subsets. Herzenberg's brilliant idea to integrate four essential 20th-century discoveries related to cellular metrics into a single platform set the stage for a new phase of complex analytical platforms to support the fight against diseases. They include multi-laser excitation, hybridoma technology for tagging fluorescently tagged monoclonal antibodies, signal processing with fast microchips and multi-channel cell sorting.</p><p>Thanks to rapid access to information, when visiting Paul Robertson's virtual library of Cytometry History at Perdue University, it is possible to appreciate how rapidly flow cytometry has matured in over five decades. In minutes, one learns about the interactions between Mack Fulwyler, Len Herzenberg, Bob Auer, Ceasar Milstein, Howard Shapiro and many other fascinating pioneers of the bio-convergence revolution of the 20th-century. The cell sorting technology uses piezo-based oscillation to charge saline droplet-enveloped cells, which are transported to be analyzed and sorted to isolate leukocyte phenotypes of interest. The droplet formation for cell sorting was Fulwyler's adaptation of technology developed for inkjet printers. With fluorescence activation, cells of interest become visible and available to be sorted for functional verification if required. In the 19th century, philosopher Arthur Schopenhauer said, “Talent hits a target no one can hit; genius hits a target no one else can see.” For a half-century, thanks to Herzenberg's contribution, most of us with talent could see leukocyte subsets with statistical significance. Steady advancements in FC continue, and multi-labeled cells can be analyzed with confidence in clinical FC laboratories with tenacity and some talent. This nostalgic indulgence is now over, and the highlights of this issue are to follow.</p><p>Three original articles and two reports on best practices are covered. The first original article is about a novel optimization method to monitor B-cell maturation antigen-targeted chimeric antigen receptors in peripheral blood. The second is an update in the understanding of the role of CD20<sup>+</sup> T-cells. The third article is about the performance of a novel 8-color panel for measurable residual disease assessment in CLL. The first best practice report validates a T-cell receptor β-chain on the constant region (TRBC) immunophenotyping protocol. This new technology improves the diagnosis of T-cell neoplasm. The other best practice report covers a more comprehensive monocyte subset analysis with flow cytometry, with increased sensitivity.</p><p>“Optimization of a flow cytometry test for routine monitoring of B-cell maturation antigen-targeted CAR in peripheral blood” is about monitoring B-cell maturation with targeted chimeric antigen T-cell receptors. The authors report encouraging news about the overall treatment success with multiple myeloma. Even on occasions when initial treatment fails, resorting to relapsed or refractory disease treatment results are encouraging with the two available FDA-licensed drugs. The authors discuss the need for a biomarker to offer a clinical outcome assessment tool. This is possible because of the availability of two reportable variables: CAR T-cell expansion kinetics with toxicity and the ability to accurately quantify B-cell maturation antigen (BCMA) CAR T-cells from peripheral blood. An earlier report from Das et al. (<span>2022</span>) indicated that immunophenotypic profile situation and post-therapy alteration in antigenic expression are available in normal, reactive, and aberrant plasma cells (NPC, RPC, and APC), respectively, for impact on measurable residual disease (MRD) assessment in multiple myeloma (MM). They concluded that antigenic aberrations on polyclonal PC signify the importance of MRD assay validation on a large cohort under normal and reactive conditions. The Das group focused on a cell-surface protein B-cell maturation antigen: CD269 as it emerged as a promising target for CAR-T cell and MAb therapies in MM. However, the knowledge of the BCMA expression pattern in myeloma patients from the Indian subcontinent was not available at that time. A study presented by Sriram et al. (<span>2022</span>) is an in-depth report of BCMA expression on abnormal plasma cells (APC) in Indian MM patients. In the UK, in a 2022 MRD assessment, MM was not incorporated into routine clinical use outside of trials. A report was released (McMillan et al., <span>2023</span>) about achieving minimal residual disease (MRD) negativity following treatment for multiple myeloma (MM) is associated with improved progression-free and overall survival. They suggest a reference MCF MM MRD method, which was stable for up to 6 days following sample collection, allowing broader access by smaller laboratories to further investigate the predictive value and clinical utility of MRD assessments.</p><p>“The role of CD20<sup>+</sup> T-cells: Insights in human peripheral blood.” This study contributes to the understanding of CD20<sup>+</sup> T-cell function in the proinflammatory cascade. The distribution of CD20<sup>−</sup> T-cells among maturation-associated T-cell compartments (naïve, central memory, transitional memory, effector memory, and effector T-cells), their polarization, activation status, and expression of immune-regulatory proteins were evaluated with flow cytometry. Their functions were assessed by measuring IFN-γ, TNF-α and IL-17 production. Compared with CD20<sup>−</sup> T-cells, CD20<sup>+</sup> T-cells represent a higher proportion of transitional memory cells. CD20<sup>+</sup> T-cells display a proinflammatory phenotype, characterized by the expansion of Th1, Th1/17, and Te1 cell subsets. They have been described and immunophenotypically characterized and purified by cell-sorting from neoplastic cells derived from lymph nodes involving T-cell/histiocyte-rich large B-cell lymphoma (Glynn &amp; Fromm, <span>2020</span>). In 2020, Gatti et al. (<span>2021</span>) published ISCCA's immunophenotypic protocol for monitoring anti-CD20 therapies in autoimmune disorders. Also, in 2020, there was a Letter to the Editor (Gadgeel et al., <span>2020</span>) about CD20<sup>+</sup> T-cells in the mediastinal mass tissue with histologically proven PMLBCL and with flow cytometric analysis. CD20<sup>+</sup> T-cells were identified as CD3, CD7, and CD2 positive cells.</p><p>“A novel 8-color FC panel for measurable residual disease (MRD) to monitor chronic lymphocytic leukemia.” The authors report on quantifiable residual disease (MRD) in chronic lymphocytic leukemia (CLL). This prognostic indicator of CLL is of interest as it has been established as predictive of progression-free survival (PFS) and overall survival (OS) in fixed-duration treatment regimens. The authors did a performance evaluation of their novel 8-color panel against the European Research Initiative on CCL (ERIC) 8-color assay. It performed well when compared with ERIC. Stage 0 hematogenous (Hgs) and their association with CD19+ Hgs in anti-CD19 therapy and conventional chemotherapy were reported (Ramalingam et al., <span>2024</span>). There is the potential to mistake a patient with residual disease if the patient received treatment that included anti-CD19 therapy as it can mimic residual Hgs. Two recent publications are addressing the use of AI to deal with the complexity of 8-color FC assay. AI analysis was used to assess measurable residual disease (Shopsowitz et al., <span>2024</span>). However, Bazinet et al. (<span>2023</span>) treated MRD after acute myeloid leukemia. They evaluated DeepFlow, an AI-assisted FC analysis software, to determine if it is suitable for performing automated clustering and identifying cell populations. They concluded that the AI-assisted analysis generates CLL MRD results similar to expert manual analysis. Their program also creates a discussion about the strengths and limitations of the AI approach.</p><p>“TRBC1 FC assay development validation and reporting considerations.” The authors reassessed the current practice of T-cell clonality by flow cytometry. They explain that because a significant gap existed between surface light chains on B-cells, for a long time it was a dependable marker for workup for hematological samples compared with T-cell oncology workups. T-cell receptor β-chain constant region (TRBC) was introduced as an alternative to TGR-V. Until recently, the assessment of clonal T-cells relied on varying marker expression levels, often providing non-specific interpretation. However, the discovery of TRBC1 puts clonal T-cell assessment by flow cytometry much closer to the clinical evaluation with B-cell clonal analysis. Recently Castillo et al. (<span>2024</span>) published a report about diagnosing T-cell non-Hodgkin lymphomas (NHL) with a MAb specific for T-cell receptor β-constant region1 (TRBC1). A study by Lee et al. (<span>2024</span>) covered an optimized whole-blood assay using human recombinant sBCMA to monitor the presence of BCMA CAR T-cells in peripheral blood. Such monitoring can be incorporated into clinical diagnostics, including the proportions of T-cells expressing the anti-BCMA CAR and absolute counts of CAR T cells/μL blood. Wang et al. (<span>2023</span>) reported a method for optimizing fluorochromes for TRBC1 to improve separation between positive and negative cells.</p><p>“Technical gating and interpretation recommendations for partitioning circulating monocyte subsets assessed by flow cytometry.” In the early days of clinical FC, monocytes were a hindrance as they had fewer CD4 MAb receptors than T-helper cells. Hence, they occasionally did compromise CD4 T-cell reporting. A second marker was added to avoid obtaining unreliable T-helper cell counts for HIV disease immunophenotyping (CD3<sup>+</sup>/CD4<sup>+</sup>). Over the past two decades, monocyte assays have been developed to detect CMML. The partitioning of circulatory monocyte subsets by flow cytometry was recommended by Wagner-Ballon et al. (<span>2023</span>). There are three monocyte subsets: cMo(CD14<sup>++</sup>/CD16<sup>−</sup>), iMo(CD14<sup>++</sup>/CD16<sup>+</sup>), and ncMo(CD14<sup>low/−</sup>/CD16<sup>+</sup>). In 2021, Devitt et al. (<span>2023</span>) disclosed the validation protocol for monocytic leukemia diagnosis at the European LeukemiaNet International MDS-Flow Cytometry Working Group (ELN iMDS-Flow). A year later, Jurado et al. (<span>2023</span>) published an optimization of monocyte gating protocol and summarized an enhanced validation approach using qualitative and semiquantitative flow cytometry technology.</p>\",\"PeriodicalId\":10883,\"journal\":{\"name\":\"Cytometry Part B: Clinical Cytometry\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.3000,\"publicationDate\":\"2024-05-30\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cyto.b.22184\",\"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.22184\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"MEDICAL LABORATORY TECHNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cytometry Part B: Clinical Cytometry","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cyto.b.22184","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

摘要

就在 50 年前的 1974 年,第一台荧光激活细胞分拣机(FACS)上市销售。Becton-Dickenson(BD)公司经斯坦福大学授权,推出了名为 FACS-1 的 FACS 分拣平台。斯坦福大学的赫岑伯格小组早在两年前就为这一新型流式细胞仪(FC)平台申请了专利。时至今日,"FACS "这一流行缩写仍被误用,因为大多数 BD FC 都是细胞分析仪,但它们都被称为 FACS 机器。无论流式细胞仪能否分拣细胞,它们都能检测白细胞亚群上与荧光标签结合的受体。赫岑伯格将 20 世纪与细胞指标相关的四项重要发现整合到一个平台上,他的这一绝妙想法为新阶段的复杂分析平台奠定了基础,为抗击疾病提供了支持。这些发现包括多激光激发、用于标记荧光标记单克隆抗体的杂交瘤技术、使用快速微芯片进行信号处理以及多通道细胞分拣。由于信息获取迅速,在访问保罗-罗伯逊位于珀杜大学的细胞测量历史虚拟图书馆时,可以了解到流式细胞仪在五十多年间是如何迅速发展成熟的。在短短几分钟内,人们就能了解到马克-福尔韦勒(Mack Fulwyler)、伦-赫岑伯格(Len Herzenberg)、鲍勃-奥尔(Bob Auer)、凯撒-米尔斯坦(Ceasar Milstein)、霍华德-夏皮罗(Howard Shapiro)以及 20 世纪生物融合革命的许多其他杰出先驱之间的互动。细胞分拣技术利用压电振荡为生理盐水液滴包被的细胞充电,这些细胞被输送到分析和分拣中心,以分离出感兴趣的白细胞表型。用于细胞分拣的液滴形成是 Fulwyler 对喷墨打印机技术的改造。通过荧光激活,感兴趣的细胞变得可见,并可根据需要进行分拣,以进行功能验证。19 世纪,哲学家亚瑟-叔本华曾说过:"天赋能击中别人无法击中的目标,而天才则能击中别人看不到的目标"。半个世纪以来,由于赫岑伯格的贡献,我们大多数有天赋的人都能看到具有统计学意义的白细胞亚群。FC技术在不断进步,临床FC实验室只要有韧劲和天赋,就能自信地分析多标记细胞。本期的精彩内容如下:三篇原创文章和两篇最佳实践报告。第一篇原创文章是关于监测外周血中 B 细胞成熟抗原靶向嵌合抗原受体的新型优化方法。第二篇是对 CD20+ T 细胞作用的最新认识。第三篇文章介绍了用于评估 CLL 可测量残留疾病的新型 8 色板的性能。第一篇最佳实践报告验证了恒定区T细胞受体β链(TRBC)免疫分型方案。这项新技术改进了 T 细胞肿瘤的诊断。另一份最佳实践报告涉及用流式细胞术进行更全面的单核细胞亚群分析,并提高了灵敏度。"优化用于常规监测外周血中B细胞成熟抗原靶向CAR的流式细胞术检测 "涉及用靶向嵌合抗原T细胞受体监测B细胞成熟。作者报告了有关多发性骨髓瘤总体治疗成功率的令人鼓舞的消息。即使在初始治疗失败的情况下,复发或难治性疾病的治疗结果也令人鼓舞。作者讨论了生物标记物提供临床结果评估工具的必要性。之所以能做到这一点,是因为有两个可报告的变量:CAR T 细胞扩增动力学与毒性,以及从外周血中准确量化 B 细胞成熟抗原 (BCMA) CAR T 细胞的能力。Das 等人(2022 年)的一份早期报告指出,正常、反应性和异常浆细胞(NPC、RPC 和 APC)的免疫表型情况和治疗后抗原表达的改变分别可对多发性骨髓瘤(MM)的可测量残留疾病(MRD)评估产生影响。他们的结论是,多克隆 PC 上的抗原畸变表明,在正常和反应性条件下对大量人群进行 MRD 检测验证非常重要。Das 小组重点研究了细胞表面蛋白 B 细胞成熟抗原:CD269是CAR-T细胞和MAb疗法治疗MM的有望靶点。然而,当时还不了解印度次大陆骨髓瘤患者的 BCMA 表达模式。Sriram 等人(2022 年)的研究深入报告了印度 MM 患者异常浆细胞(APC)中 BCMA 的表达情况。在英国,在 2022 年的 MRD 评估中,MM 未被纳入试验之外的常规临床应用。英国发布了一份报告(McMillan et al.
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Issue highlights—May 2024

Issue highlights—May 2024

Just 50 years ago, in 1974, the first fluorescence-activated cell sorter (FACS) was ready for sale. Becton-Dickenson (BD) with a license from Stanford University introduced the FACS sorting platform, which was called the FACS-1. The Herzenberg group at Stanford patented this new flow cytometry (FC) platform 2 years earlier. To this day the popular acronym “FACS” is misused in that most BD FC are cell analyzers, yet they are all called FACS machines. Whether or not a flow cytometer can sort cells, they all detect receptors bound with fluorescent tags on leukocyte subsets. Herzenberg's brilliant idea to integrate four essential 20th-century discoveries related to cellular metrics into a single platform set the stage for a new phase of complex analytical platforms to support the fight against diseases. They include multi-laser excitation, hybridoma technology for tagging fluorescently tagged monoclonal antibodies, signal processing with fast microchips and multi-channel cell sorting.

Thanks to rapid access to information, when visiting Paul Robertson's virtual library of Cytometry History at Perdue University, it is possible to appreciate how rapidly flow cytometry has matured in over five decades. In minutes, one learns about the interactions between Mack Fulwyler, Len Herzenberg, Bob Auer, Ceasar Milstein, Howard Shapiro and many other fascinating pioneers of the bio-convergence revolution of the 20th-century. The cell sorting technology uses piezo-based oscillation to charge saline droplet-enveloped cells, which are transported to be analyzed and sorted to isolate leukocyte phenotypes of interest. The droplet formation for cell sorting was Fulwyler's adaptation of technology developed for inkjet printers. With fluorescence activation, cells of interest become visible and available to be sorted for functional verification if required. In the 19th century, philosopher Arthur Schopenhauer said, “Talent hits a target no one can hit; genius hits a target no one else can see.” For a half-century, thanks to Herzenberg's contribution, most of us with talent could see leukocyte subsets with statistical significance. Steady advancements in FC continue, and multi-labeled cells can be analyzed with confidence in clinical FC laboratories with tenacity and some talent. This nostalgic indulgence is now over, and the highlights of this issue are to follow.

Three original articles and two reports on best practices are covered. The first original article is about a novel optimization method to monitor B-cell maturation antigen-targeted chimeric antigen receptors in peripheral blood. The second is an update in the understanding of the role of CD20+ T-cells. The third article is about the performance of a novel 8-color panel for measurable residual disease assessment in CLL. The first best practice report validates a T-cell receptor β-chain on the constant region (TRBC) immunophenotyping protocol. This new technology improves the diagnosis of T-cell neoplasm. The other best practice report covers a more comprehensive monocyte subset analysis with flow cytometry, with increased sensitivity.

“Optimization of a flow cytometry test for routine monitoring of B-cell maturation antigen-targeted CAR in peripheral blood” is about monitoring B-cell maturation with targeted chimeric antigen T-cell receptors. The authors report encouraging news about the overall treatment success with multiple myeloma. Even on occasions when initial treatment fails, resorting to relapsed or refractory disease treatment results are encouraging with the two available FDA-licensed drugs. The authors discuss the need for a biomarker to offer a clinical outcome assessment tool. This is possible because of the availability of two reportable variables: CAR T-cell expansion kinetics with toxicity and the ability to accurately quantify B-cell maturation antigen (BCMA) CAR T-cells from peripheral blood. An earlier report from Das et al. (2022) indicated that immunophenotypic profile situation and post-therapy alteration in antigenic expression are available in normal, reactive, and aberrant plasma cells (NPC, RPC, and APC), respectively, for impact on measurable residual disease (MRD) assessment in multiple myeloma (MM). They concluded that antigenic aberrations on polyclonal PC signify the importance of MRD assay validation on a large cohort under normal and reactive conditions. The Das group focused on a cell-surface protein B-cell maturation antigen: CD269 as it emerged as a promising target for CAR-T cell and MAb therapies in MM. However, the knowledge of the BCMA expression pattern in myeloma patients from the Indian subcontinent was not available at that time. A study presented by Sriram et al. (2022) is an in-depth report of BCMA expression on abnormal plasma cells (APC) in Indian MM patients. In the UK, in a 2022 MRD assessment, MM was not incorporated into routine clinical use outside of trials. A report was released (McMillan et al., 2023) about achieving minimal residual disease (MRD) negativity following treatment for multiple myeloma (MM) is associated with improved progression-free and overall survival. They suggest a reference MCF MM MRD method, which was stable for up to 6 days following sample collection, allowing broader access by smaller laboratories to further investigate the predictive value and clinical utility of MRD assessments.

“The role of CD20+ T-cells: Insights in human peripheral blood.” This study contributes to the understanding of CD20+ T-cell function in the proinflammatory cascade. The distribution of CD20 T-cells among maturation-associated T-cell compartments (naïve, central memory, transitional memory, effector memory, and effector T-cells), their polarization, activation status, and expression of immune-regulatory proteins were evaluated with flow cytometry. Their functions were assessed by measuring IFN-γ, TNF-α and IL-17 production. Compared with CD20 T-cells, CD20+ T-cells represent a higher proportion of transitional memory cells. CD20+ T-cells display a proinflammatory phenotype, characterized by the expansion of Th1, Th1/17, and Te1 cell subsets. They have been described and immunophenotypically characterized and purified by cell-sorting from neoplastic cells derived from lymph nodes involving T-cell/histiocyte-rich large B-cell lymphoma (Glynn & Fromm, 2020). In 2020, Gatti et al. (2021) published ISCCA's immunophenotypic protocol for monitoring anti-CD20 therapies in autoimmune disorders. Also, in 2020, there was a Letter to the Editor (Gadgeel et al., 2020) about CD20+ T-cells in the mediastinal mass tissue with histologically proven PMLBCL and with flow cytometric analysis. CD20+ T-cells were identified as CD3, CD7, and CD2 positive cells.

“A novel 8-color FC panel for measurable residual disease (MRD) to monitor chronic lymphocytic leukemia.” The authors report on quantifiable residual disease (MRD) in chronic lymphocytic leukemia (CLL). This prognostic indicator of CLL is of interest as it has been established as predictive of progression-free survival (PFS) and overall survival (OS) in fixed-duration treatment regimens. The authors did a performance evaluation of their novel 8-color panel against the European Research Initiative on CCL (ERIC) 8-color assay. It performed well when compared with ERIC. Stage 0 hematogenous (Hgs) and their association with CD19+ Hgs in anti-CD19 therapy and conventional chemotherapy were reported (Ramalingam et al., 2024). There is the potential to mistake a patient with residual disease if the patient received treatment that included anti-CD19 therapy as it can mimic residual Hgs. Two recent publications are addressing the use of AI to deal with the complexity of 8-color FC assay. AI analysis was used to assess measurable residual disease (Shopsowitz et al., 2024). However, Bazinet et al. (2023) treated MRD after acute myeloid leukemia. They evaluated DeepFlow, an AI-assisted FC analysis software, to determine if it is suitable for performing automated clustering and identifying cell populations. They concluded that the AI-assisted analysis generates CLL MRD results similar to expert manual analysis. Their program also creates a discussion about the strengths and limitations of the AI approach.

“TRBC1 FC assay development validation and reporting considerations.” The authors reassessed the current practice of T-cell clonality by flow cytometry. They explain that because a significant gap existed between surface light chains on B-cells, for a long time it was a dependable marker for workup for hematological samples compared with T-cell oncology workups. T-cell receptor β-chain constant region (TRBC) was introduced as an alternative to TGR-V. Until recently, the assessment of clonal T-cells relied on varying marker expression levels, often providing non-specific interpretation. However, the discovery of TRBC1 puts clonal T-cell assessment by flow cytometry much closer to the clinical evaluation with B-cell clonal analysis. Recently Castillo et al. (2024) published a report about diagnosing T-cell non-Hodgkin lymphomas (NHL) with a MAb specific for T-cell receptor β-constant region1 (TRBC1). A study by Lee et al. (2024) covered an optimized whole-blood assay using human recombinant sBCMA to monitor the presence of BCMA CAR T-cells in peripheral blood. Such monitoring can be incorporated into clinical diagnostics, including the proportions of T-cells expressing the anti-BCMA CAR and absolute counts of CAR T cells/μL blood. Wang et al. (2023) reported a method for optimizing fluorochromes for TRBC1 to improve separation between positive and negative cells.

“Technical gating and interpretation recommendations for partitioning circulating monocyte subsets assessed by flow cytometry.” In the early days of clinical FC, monocytes were a hindrance as they had fewer CD4 MAb receptors than T-helper cells. Hence, they occasionally did compromise CD4 T-cell reporting. A second marker was added to avoid obtaining unreliable T-helper cell counts for HIV disease immunophenotyping (CD3+/CD4+). Over the past two decades, monocyte assays have been developed to detect CMML. The partitioning of circulatory monocyte subsets by flow cytometry was recommended by Wagner-Ballon et al. (2023). There are three monocyte subsets: cMo(CD14++/CD16), iMo(CD14++/CD16+), and ncMo(CD14low/−/CD16+). In 2021, Devitt et al. (2023) disclosed the validation protocol for monocytic leukemia diagnosis at the European LeukemiaNet International MDS-Flow Cytometry Working Group (ELN iMDS-Flow). A year later, Jurado et al. (2023) published an optimization of monocyte gating protocol and summarized an enhanced validation approach using qualitative and semiquantitative flow cytometry technology.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
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