异基因造血干细胞移植过程中人I型T淋巴细胞病毒向供体来源T细胞的传播

IF 1.3 Q4 HEMATOLOGY
M. Hirosawa, D. Niino, J. Tsukada
{"title":"异基因造血干细胞移植过程中人I型T淋巴细胞病毒向供体来源T细胞的传播","authors":"M. Hirosawa, D. Niino, J. Tsukada","doi":"10.14740/jh973","DOIUrl":null,"url":null,"abstract":"Human T-lymphotropic virus type I (HTLV-I) is a retrovirus that causes adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4-positive mature T-cell malignancy with a dismal prognosis [1, 2]. In addition, the virus is associated with chronic inflammatory diseases such as HTLV-I-associated myelopathy (HAM)/tropical spastic paraparesis (TSP), arthritis, uveitis, dermatitis, and bronchioloalveolar disorders. It is estimated that HTLV-I infects 5 20 million individuals worldwide [3, 4]. HTLV-I is transmitted via breast feeding, sexual intercourse, needle sharing, and blood products containing cells. Organ transplantation has also been considered a rare route of HTLV-I transmission [5]. Here, we describe a case of HTLV-I transmission to T cells of an HTLV-I-negative donor through allogeneic hematopoietic stem cell transplantation (allo-HSCT). A 56-year-old man with a history of ATLL was referred to our hospital for dyspnea. Two years before the presentation, he underwent allogeneic peripheral blood stem cell transplantation (allo-PBSCT) from an HTLV-I-negative female donor with a reduced intensity conditioning (RIC) regimen consisting of fludarabine, melphalan, and total body irradiation (4 Gy). T-cell depletion was carried out using thymoglobulin. At the time of transplantation, the patient had systemic lymphadenopathy due to refractory ATLL. After transplantation, the patient achieved a complete remission. The HTLV-I-negative donor had neither atypical lymphocytes in the peripheral blood (PB) nor lymphadenopathy. The seronegativity of anti-HTLV-I antibody was confirmed by chemiluminescence enzyme immunoassay. Serum immunoglobulin levels of the donor were within normal limits. When the patient was referred to our hospital with dyspnea, he was receiving tacrolimus therapy for chronic graft-vshost disease (GVHD). He immediately underwent a complete blood count, which revealed 11 × 109/L white blood cells with 17% of atypical lymphocytes. The morphologic feature of the atypical lymphocytes was pleomorphic with condensed chromatin and convoluted or lobulated nucleus (Fig. 1a). Flow cytometric analysis (FCM) revealed that the atypical lymphocytes were positive for CD3 (Fig. 1b), CD25 (Fig. 1b) and CD4, and negative for CD8. Furthermore, the atypical lymphocytes, unlike his pretransplant ATLL cells, expressed CD7 (Fig. 1c). An increased HTLV-I proviral load (PVL) of 397.2 copies/1,000 PB mononuclear cells (PBMCs) was detected. Although no hypercalcemia was observed, serum lactate dehydrogenase and soluble interleukin (IL)-2 receptor levels were elevated to 290 U/L (normal: 124 222 U/L) and 30,573 U/ mL (normal: 145 519 U/mL), respectively. Epstein-Barr virus (EBV) and cytomegalovirus were negative in real-time quantitative polymerase chain reaction. Computed tomography of the chest revealed a mediastinal tumor and pleural effusion (arrows: Fig. 1d). Tumor biopsy showed diffuse infiltration of medium to large, atypical cells with irregular nuclear contour (Fig. 1e). The infiltrating tumor cells showed strong immunohistochemical staining for CD3 (Fig. 1f), CD4 (Fig. 1g), CD7 (Fig. 1h), and CD25 (Fig. 1i), but they were negative for CD8 (Fig. 1j) and EBV-encoded small RNA. FCM of the infiltrating tumor cells showed the same results as those of PB. Southern blot analysis (SBA) of the atypical lymphocytes using HTLV-I DNA as a probe (Fig. 2a) revealed internal bands following Pst1 digestion (lane 6; arrowheads). However, no monoclonal band pattern was observed in EcoR1 digestion (lane 5; arrow). In contrast, SBA results obtained from his pretransplant ATLL cells (Fig. 2b) revealed monoclonal integration of a defective HTLV-I provirus (lane 5; arrow). Moreover, XY-fluorescence in situ hybridization (XY-FISH) of the atypical lymphocytes was performed to determine the origin of the atypical lymphocytes. All tested cells revealed a typical XX pattern of a female donor (Fig. 2c). The G-band karyotype of the lymphocytes was also 46, XX. Bone marrow examination confirmed complete donor chimerism. Thus, our results demonstrated acquired HTLV-I infection of donor-origin T cells in the recipient. Two months after the discontinuation of tacrolimus, white blood cell counts in PB returned to normal, and no atypical lymManuscript submitted January 10, 2022, accepted February 15, 2022 Published online March 12, 2022","PeriodicalId":15964,"journal":{"name":"Journal of hematology","volume":null,"pages":null},"PeriodicalIF":1.3000,"publicationDate":"2022-03-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"1","resultStr":"{\"title\":\"Transmission of Human T-Lymphotropic Virus Type I to Donor-Origin T Cells During Allogeneic Hematopoietic Stem Cell Transplantation\",\"authors\":\"M. Hirosawa, D. Niino, J. Tsukada\",\"doi\":\"10.14740/jh973\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Human T-lymphotropic virus type I (HTLV-I) is a retrovirus that causes adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4-positive mature T-cell malignancy with a dismal prognosis [1, 2]. In addition, the virus is associated with chronic inflammatory diseases such as HTLV-I-associated myelopathy (HAM)/tropical spastic paraparesis (TSP), arthritis, uveitis, dermatitis, and bronchioloalveolar disorders. It is estimated that HTLV-I infects 5 20 million individuals worldwide [3, 4]. HTLV-I is transmitted via breast feeding, sexual intercourse, needle sharing, and blood products containing cells. Organ transplantation has also been considered a rare route of HTLV-I transmission [5]. Here, we describe a case of HTLV-I transmission to T cells of an HTLV-I-negative donor through allogeneic hematopoietic stem cell transplantation (allo-HSCT). A 56-year-old man with a history of ATLL was referred to our hospital for dyspnea. Two years before the presentation, he underwent allogeneic peripheral blood stem cell transplantation (allo-PBSCT) from an HTLV-I-negative female donor with a reduced intensity conditioning (RIC) regimen consisting of fludarabine, melphalan, and total body irradiation (4 Gy). T-cell depletion was carried out using thymoglobulin. At the time of transplantation, the patient had systemic lymphadenopathy due to refractory ATLL. After transplantation, the patient achieved a complete remission. The HTLV-I-negative donor had neither atypical lymphocytes in the peripheral blood (PB) nor lymphadenopathy. The seronegativity of anti-HTLV-I antibody was confirmed by chemiluminescence enzyme immunoassay. Serum immunoglobulin levels of the donor were within normal limits. When the patient was referred to our hospital with dyspnea, he was receiving tacrolimus therapy for chronic graft-vshost disease (GVHD). He immediately underwent a complete blood count, which revealed 11 × 109/L white blood cells with 17% of atypical lymphocytes. The morphologic feature of the atypical lymphocytes was pleomorphic with condensed chromatin and convoluted or lobulated nucleus (Fig. 1a). Flow cytometric analysis (FCM) revealed that the atypical lymphocytes were positive for CD3 (Fig. 1b), CD25 (Fig. 1b) and CD4, and negative for CD8. Furthermore, the atypical lymphocytes, unlike his pretransplant ATLL cells, expressed CD7 (Fig. 1c). An increased HTLV-I proviral load (PVL) of 397.2 copies/1,000 PB mononuclear cells (PBMCs) was detected. Although no hypercalcemia was observed, serum lactate dehydrogenase and soluble interleukin (IL)-2 receptor levels were elevated to 290 U/L (normal: 124 222 U/L) and 30,573 U/ mL (normal: 145 519 U/mL), respectively. Epstein-Barr virus (EBV) and cytomegalovirus were negative in real-time quantitative polymerase chain reaction. Computed tomography of the chest revealed a mediastinal tumor and pleural effusion (arrows: Fig. 1d). Tumor biopsy showed diffuse infiltration of medium to large, atypical cells with irregular nuclear contour (Fig. 1e). The infiltrating tumor cells showed strong immunohistochemical staining for CD3 (Fig. 1f), CD4 (Fig. 1g), CD7 (Fig. 1h), and CD25 (Fig. 1i), but they were negative for CD8 (Fig. 1j) and EBV-encoded small RNA. FCM of the infiltrating tumor cells showed the same results as those of PB. Southern blot analysis (SBA) of the atypical lymphocytes using HTLV-I DNA as a probe (Fig. 2a) revealed internal bands following Pst1 digestion (lane 6; arrowheads). However, no monoclonal band pattern was observed in EcoR1 digestion (lane 5; arrow). In contrast, SBA results obtained from his pretransplant ATLL cells (Fig. 2b) revealed monoclonal integration of a defective HTLV-I provirus (lane 5; arrow). Moreover, XY-fluorescence in situ hybridization (XY-FISH) of the atypical lymphocytes was performed to determine the origin of the atypical lymphocytes. All tested cells revealed a typical XX pattern of a female donor (Fig. 2c). The G-band karyotype of the lymphocytes was also 46, XX. Bone marrow examination confirmed complete donor chimerism. Thus, our results demonstrated acquired HTLV-I infection of donor-origin T cells in the recipient. Two months after the discontinuation of tacrolimus, white blood cell counts in PB returned to normal, and no atypical lymManuscript submitted January 10, 2022, accepted February 15, 2022 Published online March 12, 2022\",\"PeriodicalId\":15964,\"journal\":{\"name\":\"Journal of hematology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":1.3000,\"publicationDate\":\"2022-03-12\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"1\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal of hematology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.14740/jh973\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q4\",\"JCRName\":\"HEMATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of hematology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.14740/jh973","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q4","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 1

摘要

人类嗜T淋巴细胞病毒I型(HTLV-I)是一种逆转录病毒,可导致成人T细胞白血病/淋巴瘤(ATLL),这是一种侵袭性CD4阳性的成熟T细胞恶性肿瘤,预后不佳[1,2]。此外,该病毒与慢性炎症性疾病有关,如HTLV-I相关脊髓病(HAM)/热带痉挛性轻瘫(TSP)、关节炎、葡萄膜炎、皮炎和细支气管肺泡疾病。据估计,HTLV-I在全球范围内感染了5200万人[3,4]。HTLV-I通过母乳喂养、性交、共用针头和含有细胞的血液制品传播。器官移植也被认为是HTLV-I传播的一种罕见途径[5]。在此,我们描述了一例通过异基因造血干细胞移植(allo-HSCT)将HTLV-I传递给HTLV-I阴性供体的T细胞的病例。一名有ATLL病史的56岁男子因呼吸困难被转诊到我们医院。在陈述前两年,他接受了来自HTLV-I阴性女性供体的异基因外周血干细胞移植(allo-PBSCT),采用由氟达拉滨、美法仑和全身照射(4Gy)组成的低强度调节(RIC)方案。使用胸腺球蛋白进行T细胞耗竭。移植时,患者因难治性ATLL而出现全身淋巴结病。移植后,病人病情完全缓解。HTLV-I阴性供体的外周血(PB)中既没有非典型淋巴细胞,也没有淋巴结病。化学发光酶免疫测定法证实了抗HTLV-I抗体的血清阴性。捐献者的血清免疫球蛋白水平在正常范围内。当患者因呼吸困难被转诊到我们医院时,他正在接受他克莫司治疗慢性移植物抗宿主病(GVHD)。他立即接受了全面的血液计数,结果显示11×109/L的白细胞和17%的非典型淋巴细胞。非典型淋巴细胞的形态学特征为多形性,染色质浓缩,细胞核卷曲或分叶(图第1a段)。流式细胞术分析(FCM)显示,非典型淋巴细胞CD3阳性(图1b)、CD25阳性(图1b)和CD4,CD8阴性。此外,非典型淋巴细胞不同于他的移植前ATLL细胞,表达CD7(图1c)。检测到397.2拷贝/1000 PB单核细胞(PBMC)的HTLV-I前病毒载量(PVL)增加。尽管未观察到高钙血症,但血清乳酸脱氢酶和可溶性白细胞介素(IL)-2受体水平分别升高至290 U/L(正常:124 222 U/L)和30573 U/mL(正常:145 519 U/mL)。EB病毒和巨细胞病毒实时定量聚合酶链反应均为阴性。胸部计算机断层扫描显示纵隔肿瘤和胸腔积液(箭头:图1d)。肿瘤活检显示中到大的弥漫性浸润,非典型细胞具有不规则的细胞核轮廓(图1e)。浸润的肿瘤细胞显示出CD3的强免疫组织化学染色(图1f)、CD4(图1g)、CD7(图1h)和CD25(图1i),但它们对CD8呈阴性(图1j)和EBV编码的小RNA。对浸润性肿瘤细胞进行FCM检测,结果与PB相同。使用HTLV-I DNA作为探针对非典型淋巴细胞进行Southern印迹分析(SBA)(图2a)显示Pst1消化后的内部条带(泳道6;箭头)。然而,在EcoR1消化中没有观察到单克隆带模式(泳道5;箭头)。相反,从他的移植前ATLL细胞获得的SBA结果(图2b)揭示了有缺陷的HTLV-I前病毒的单克隆整合(泳道5;箭头)。此外,对非典型淋巴细胞进行XY荧光原位杂交(XY-FISH)以确定非典型淋巴细胞的起源。所有测试的细胞都显示出女性供体的典型XX模式(图2c)。淋巴细胞的G带核型也为46,XX。骨髓检查证实了完全的供体嵌合。因此,我们的结果证明了受体中供体来源的T细胞的获得性HTLV-I感染。他克莫司停药两个月后,PB中的白细胞计数恢复正常,没有非典型lymManuscript于2022年1月10日提交,于2022年2月15日接受。于2022年3月12日在线发布
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Transmission of Human T-Lymphotropic Virus Type I to Donor-Origin T Cells During Allogeneic Hematopoietic Stem Cell Transplantation
Human T-lymphotropic virus type I (HTLV-I) is a retrovirus that causes adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4-positive mature T-cell malignancy with a dismal prognosis [1, 2]. In addition, the virus is associated with chronic inflammatory diseases such as HTLV-I-associated myelopathy (HAM)/tropical spastic paraparesis (TSP), arthritis, uveitis, dermatitis, and bronchioloalveolar disorders. It is estimated that HTLV-I infects 5 20 million individuals worldwide [3, 4]. HTLV-I is transmitted via breast feeding, sexual intercourse, needle sharing, and blood products containing cells. Organ transplantation has also been considered a rare route of HTLV-I transmission [5]. Here, we describe a case of HTLV-I transmission to T cells of an HTLV-I-negative donor through allogeneic hematopoietic stem cell transplantation (allo-HSCT). A 56-year-old man with a history of ATLL was referred to our hospital for dyspnea. Two years before the presentation, he underwent allogeneic peripheral blood stem cell transplantation (allo-PBSCT) from an HTLV-I-negative female donor with a reduced intensity conditioning (RIC) regimen consisting of fludarabine, melphalan, and total body irradiation (4 Gy). T-cell depletion was carried out using thymoglobulin. At the time of transplantation, the patient had systemic lymphadenopathy due to refractory ATLL. After transplantation, the patient achieved a complete remission. The HTLV-I-negative donor had neither atypical lymphocytes in the peripheral blood (PB) nor lymphadenopathy. The seronegativity of anti-HTLV-I antibody was confirmed by chemiluminescence enzyme immunoassay. Serum immunoglobulin levels of the donor were within normal limits. When the patient was referred to our hospital with dyspnea, he was receiving tacrolimus therapy for chronic graft-vshost disease (GVHD). He immediately underwent a complete blood count, which revealed 11 × 109/L white blood cells with 17% of atypical lymphocytes. The morphologic feature of the atypical lymphocytes was pleomorphic with condensed chromatin and convoluted or lobulated nucleus (Fig. 1a). Flow cytometric analysis (FCM) revealed that the atypical lymphocytes were positive for CD3 (Fig. 1b), CD25 (Fig. 1b) and CD4, and negative for CD8. Furthermore, the atypical lymphocytes, unlike his pretransplant ATLL cells, expressed CD7 (Fig. 1c). An increased HTLV-I proviral load (PVL) of 397.2 copies/1,000 PB mononuclear cells (PBMCs) was detected. Although no hypercalcemia was observed, serum lactate dehydrogenase and soluble interleukin (IL)-2 receptor levels were elevated to 290 U/L (normal: 124 222 U/L) and 30,573 U/ mL (normal: 145 519 U/mL), respectively. Epstein-Barr virus (EBV) and cytomegalovirus were negative in real-time quantitative polymerase chain reaction. Computed tomography of the chest revealed a mediastinal tumor and pleural effusion (arrows: Fig. 1d). Tumor biopsy showed diffuse infiltration of medium to large, atypical cells with irregular nuclear contour (Fig. 1e). The infiltrating tumor cells showed strong immunohistochemical staining for CD3 (Fig. 1f), CD4 (Fig. 1g), CD7 (Fig. 1h), and CD25 (Fig. 1i), but they were negative for CD8 (Fig. 1j) and EBV-encoded small RNA. FCM of the infiltrating tumor cells showed the same results as those of PB. Southern blot analysis (SBA) of the atypical lymphocytes using HTLV-I DNA as a probe (Fig. 2a) revealed internal bands following Pst1 digestion (lane 6; arrowheads). However, no monoclonal band pattern was observed in EcoR1 digestion (lane 5; arrow). In contrast, SBA results obtained from his pretransplant ATLL cells (Fig. 2b) revealed monoclonal integration of a defective HTLV-I provirus (lane 5; arrow). Moreover, XY-fluorescence in situ hybridization (XY-FISH) of the atypical lymphocytes was performed to determine the origin of the atypical lymphocytes. All tested cells revealed a typical XX pattern of a female donor (Fig. 2c). The G-band karyotype of the lymphocytes was also 46, XX. Bone marrow examination confirmed complete donor chimerism. Thus, our results demonstrated acquired HTLV-I infection of donor-origin T cells in the recipient. Two months after the discontinuation of tacrolimus, white blood cell counts in PB returned to normal, and no atypical lymManuscript submitted January 10, 2022, accepted February 15, 2022 Published online March 12, 2022
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
Journal of hematology
Journal of hematology HEMATOLOGY-
自引率
0.00%
发文量
29
×
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