51
|
van der Maas NG, Berghuis D, van der Burg M, Lankester AC. B Cell Reconstitution and Influencing Factors After Hematopoietic Stem Cell Transplantation in Children. Front Immunol 2019; 10:782. [PMID: 31031769 PMCID: PMC6473193 DOI: 10.3389/fimmu.2019.00782] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/25/2019] [Indexed: 12/20/2022] Open
Abstract
B cell reconstitution after hematopoietic stem cell transplantation (HSCT) is variable and influenced by different patient, donor, and treatment related factors. In this review we describe B cell reconstitution after pediatric allogeneic HST, including the kinetics of reconstitution of the different B cell subsets and the development of the B cell repertoire, and discuss the influencing factors. Observational studies show important roles for stem cell source, conditioning regimen, and graft vs. host disease in B cell reconstitution. In addition, B cell recovery can play an important role in post-transplant infections and vaccine responses to encapsulated bacteria, such as pneumococcus. A substantial number of patients experience impaired B cell function and/or dependency on Ig substitution after allogeneic HSCT. The underlying mechanisms are largely unresolved. The integrated aspects of B cell recovery after HSCT, especially BCR repertoire reconstitution, are awaiting further investigation using modern techniques in order to gain more insight into B cell reconstitution and to develop strategies to improve humoral immunity after allogeneic HSCT.
Collapse
Affiliation(s)
- Nicolaas G van der Maas
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Dagmar Berghuis
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Mirjam van der Burg
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Arjan C Lankester
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
52
|
Chen XM, Weng JY, Lai PL, Wang YL, Huang X, Geng SX, Guo LY, Huang T, Zeng LJ, Du X. [Artesunate attenuate chronic graft-versus-host disease by regulating Th17/Treg balance]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:63-68. [PMID: 30704231 PMCID: PMC7351699 DOI: 10.3760/cma.j.issn.0253-2727.2019.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
目的 探讨青蒿琥酯抗慢性移植物抗宿主病(cGVHD)的作用及可能机制。 方法 将B10D2小鼠的骨髓细胞和脾脏细胞混合悬液通过尾静脉输入近交系雌性BALB/c小鼠体内建立cGVHD模型,分别予青蒿琥酯(实验组)和丙酮腹腔(对照组)注射治疗。观察两组小鼠的cGVHD临床表现、生存时间和组织病理学改变;应用流式细胞术分析小鼠外周血和脾脏Th17和Treg细胞比例;免疫磁珠分选BALB/c小鼠脾脏CD4+T细胞,分别予丙酮和青蒿琥酯进行干预72 h,流式细胞术分析两组Th17/Treg细胞比例。 结果 ①实验组小鼠cGVHD临床症状较对照组明显减轻,生存时间延长[(55.71±6.99)d对(46.57±7.83)d,χ2=5.457,P=0.020]。②实验组小鼠皮肤及肺脏cGVHD病理损伤较轻。③与对照组比较,实验组小鼠外周血、脾脏Th17细胞比例均降低[(0.58±0.19)%对(1.51±0.18)%,t=7.233,P<0.001;(0.71±0.18)%对(1.48±0.38)%,t=3.653,P=0.011],Treg细胞比例增高[(8.40±0.23)%对(4.45±0.04)%,t=15.680,P<0.001;(10.48±0.48)%对(6.62±0.24)%,t=6.590,P=0.003],Th17/Treg细胞比值均下降(0.09±0.03对0.34±0.05,t=7.621,P=0.002;0.06±0.02对0.19±0.03,t=6.993,P=0.002)。④体外培养小鼠脾脏CD4+T细胞,青蒿琥酯干预组较对照组Th17细胞比例减低[(0.82±0.37)%对(3.39±1.22)%,t=4.044,P=0.007],Treg细胞比例明显增高[(34.63±1.29)%对(14.28±1.69)%,t=19.119,P<0.001],Th17/Treg细胞比值下降(0.24±0.09对0.02±0.01,t=4.780,P=0.003)。 结论 青蒿琥酯可通过减少Th17细胞及增加Treg细胞,恢复Th17/Treg平衡,减轻cGVHD的临床与病理学损伤,从而发挥抗cGVHD作用。
Collapse
Affiliation(s)
- X M Chen
- Department of Hematology, Guangdong General Hospital/Guangdong Academy of Medical Sciences, Guangdong Geriatrics Institute, Guangzhou 510080, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Graft-versus-host disease in recipients of male unrelated donor compared with parous female sibling donor transplants. Blood Adv 2019; 2:1022-1031. [PMID: 29739773 DOI: 10.1182/bloodadvances.2017013052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/26/2018] [Indexed: 12/18/2022] Open
Abstract
Optimal donor selection is critical for successful allogeneic hematopoietic cell transplantation (HCT). Donor sex and parity are well-established risk factors for graft-versus-host disease (GVHD), with male donors typically associated with lower rates of GVHD. Well-matched unrelated donors (URDs) have also been associated with increased risks of GVHD as compared with matched sibling donors. These observations raise the question of whether male URDs would lead to more (or less) favorable transplant outcomes as compared with parous female sibling donors. We used the Center for International Blood and Marrow Transplant Research registry to complete a retrospective cohort study in adults with acute myeloid leukemia, acute lymphoblastic leukemia, or myelodysplastic syndrome, who underwent T-cell replete HCT from these 2 donor types (parous female sibling or male URD) between 2000 and 2012. Primary outcomes included grade 2 to 4 acute GVHD (aGVHD), chronic GVHD (cGVHD), and overall survival. Secondary outcomes included disease-free survival, transplant-related mortality, and relapse. In 2813 recipients, patients receiving male URD transplants (n = 1921) had 1.6 times higher risk of grade 2 to 4 aGVHD (P < .0001). For cGVHD, recipient sex was a significant factor, so donor/recipient pairs were evaluated. Female recipients of male URD grafts had a higher risk of cGVHD than those receiving parous female sibling grafts (relative risk [RR] = 1.43, P < .0001), whereas male recipients had similar rates of cGVHD regardless of donor type (RR = 1.09, P = .23). Donor type did not significantly affect any other end point. We conclude that when available, parous female siblings are preferred over male URDs.
Collapse
|
54
|
Paul J, Nakasone H, Sahaf B, Wu F, Wang K, Ho V, Wu J, Kim H, Blazar B, Ritz J, Howard A, Cutler C, Miklos D. A confirmation of chronic graft- versus-host disease prediction using allogeneic HY antibodies following sex-mismatched hematopoietic cell transplantation. Haematologica 2019; 104:e314-e317. [PMID: 30655371 DOI: 10.3324/haematol.2018.199646] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Jed Paul
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Hideki Nakasone
- Division of Hematology, Saitama Medical Center, Jichi Medicial University, Saitama, Japan
| | - Bita Sahaf
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Fang Wu
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathy Wang
- Division of Hematological Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Vincent Ho
- Division of Hematological Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Juan Wu
- The Emmes Corporation, Rockville, MD, USA
| | - Haesook Kim
- Division of Hematological Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Bruce Blazar
- Division of Pediatrics BMT, Minneapolis, MN, USA
| | - Jerome Ritz
- Division of Hematological Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alan Howard
- Center for International Blood and Marrow Transplant Research, Minneapolis Campus, Minneapolis, MN, USA
| | - Corey Cutler
- Division of Hematological Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Miklos
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
55
|
Li X, Gao Q, Feng Y, Zhang X. Developing role of B cells in the pathogenesis and treatment of chronic GVHD. Br J Haematol 2018; 184:323-336. [PMID: 30585319 PMCID: PMC6590173 DOI: 10.1111/bjh.15719] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is a major complication affecting the long-term survival of patients after allogeneic haematopoietic stem cell transplantation. The mechanism of cGVHD is unclear, and while previous studies have primarily focused on T cells, the role of B cells in the pathogenesis of cGVHD has been less reported. However, current studies on cGVHD are increasingly focused on the important role of B cells. In this review, we will introduce the newest studies and examine the role of B cells in cGVHD in detail with respect to the following aspects: altered B cell subpopulations, aberrant B cell signalling pathways, autoantibodies and T-B cell interactions. Treatment strategies for the targeting of B cells during cGVHD will also be discussed.
Collapse
Affiliation(s)
- Xiaoping Li
- Department of Haematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qiangguo Gao
- Department of Cell Biology College of Basic Medicine, Third Military Medicine University, Chongqing, China
| | - Yimei Feng
- Department of Haematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xi Zhang
- Department of Haematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
56
|
Kuba A, Raida L. Graft versus Host Disease: From Basic Pathogenic Principles to DNA Damage Response and Cellular Senescence. Mediators Inflamm 2018; 2018:9451950. [PMID: 29785172 PMCID: PMC5896258 DOI: 10.1155/2018/9451950] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/12/2018] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Graft versus host disease (GVHD), a severe immunogenic complication of allogeneic hematopoietic stem cell transplantation (HSCT), represents the most frequent cause of transplant-related mortality (TRM). Despite a huge progress in HSCT techniques and posttransplant care, GVHD remains a significant obstacle in successful HSCT outcome. This review presents a complex summary of GVHD pathogenesis with focus on references considering basic biological processes such as DNA damage response and cellular senescence.
Collapse
Affiliation(s)
- Adam Kuba
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Ludek Raida
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
57
|
Geneugelijk K, Spierings E. Matching donor and recipient based on predicted indirectly recognizable human leucocyte antigen epitopes. Int J Immunogenet 2018; 45:41-53. [PMID: 29464898 DOI: 10.1111/iji.12359] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/21/2017] [Accepted: 02/01/2018] [Indexed: 12/27/2022]
Abstract
The predicted indirectly recognizable human leucocyte antigen (HLA) epitopes (PIRCHE) algorithm is a novel in silico algorithm to determine donor-recipient compatibility. The PIRCHE algorithm determines donor-recipient compatibility by counting the number of mismatched HLA-derived epitopes that are involved in indirect T-cell alloimmune responses; these epitopes are designated as PIRCHE. Over the last few years, the PIRCHE algorithm has been investigated in both hematopoietic stem cell transplantation and solid organ transplantation. This review describes the theory of the algorithm, its application in transplantation, and highlights the future perspectives on the clinical application of the PIRCHE algorithm.
Collapse
Affiliation(s)
- K Geneugelijk
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - E Spierings
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
58
|
B-cell targeting in chronic graft-versus-host disease. Blood 2018; 131:1399-1405. [PMID: 29437591 DOI: 10.1182/blood-2017-11-784017] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/25/2018] [Indexed: 01/08/2023] Open
Abstract
Over the last decade, our understanding of the pathophysiology of chronic graft-versus-host disease (cGVHD) has improved considerably. In this spotlight, we discuss emerging insights into the pathophysiology of cGVHD with a focus on B cells. First, we summarize supporting evidence derived from mouse and human studies. Next, novel cGVHD therapy approaches that target B cells will be covered to provide treating physicians with an overview of the rationale behind the emerging armamentarium against cGVHD.
Collapse
|
59
|
Epperla N, Ahn KW, Armand P, Jaglowski S, Ahmed S, Kenkre VP, Savani B, Jagasia M, Shah NN, Fenske TS, Sureda A, Smith SM, Hamadani M. Fludarabine and Busulfan versus Fludarabine, Cyclophosphamide, and Rituximab as Reduced-Intensity Conditioning for Allogeneic Transplantation in Follicular Lymphoma. Biol Blood Marrow Transplant 2018; 24:78-85. [PMID: 29032272 PMCID: PMC5743624 DOI: 10.1016/j.bbmt.2017.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/05/2017] [Indexed: 01/31/2023]
Abstract
Large, multicenter studies comparing commonly used reduced-intensity conditioning (RIC) approaches in follicular lymphoma (FL) have not been performed. Using the Center for International Blood and Marrow Transplant Research database, we report the outcomes of the 2 most commonly used RIC approaches, fludarabine and busulfan (Flu/Bu) versus fludarabine, cyclophosphamide, and rituximab (FCR) in FL patients. We evaluated 200 FL patients undergoing allogeneic hematopoietic cell transplantation (allo-HCT) who received RIC with either Flu/Bu (n = 98) or FCR (n = 102) during 2008 to 2014. All patients received peripheral blood grafts, and graft-versus-host disease (GVHD) prophylaxis was limited to calcineurin inhibitor-based approaches. Median follow-up of survivors in the Flu/Bu and FCR groups was 48 months and 46 months, respectively. On univariate analysis in the Flu/Bu and FCR groups, the 3-year rates of nonrelapse mortality (11% versus 11%, P = .94), relapse/progression (18% versus 15%, P = .54), progression-free survival (PFS) (71% versus 74%, P = .65), and overall survival (OS) (73% versus 81%, P = .18) were not significantly different. On multivariate analysis no difference was seen between the FCR and Flu/Bu cohorts in terms of grades II to IV (relative risk [RR], 1.06; 95% confidence interval [CI], .59 to 1.93; P = .84) or grades III to IV (RR, 1.18; 95% CI, .47 to 2.99; P = .72) acute GVHD, nonrelapse mortality (RR, .83; 95% CI, .38 to 1.82; P = .64), relapse/progression (RR, .99; 95% CI, .49 to 1.98; P = .97), PFS (RR, .92; 95% CI, .55 to 1.54; P = .76), or OS (RR, .70; 95% CI, .40 to 1.23; P = .21) risk. However, RIC with FCR was associated with a significantly reduced chronic GVHD risk (RR, .52; 95% CI, .36 to .77; P = .001). RIC with either Flu/Bu or FCR in patients with FL undergoing allo-HCT provides excellent 3-year OS, with acceptable rates of nonrelapse mortality. FCR-based conditioning was associated with a lower risk of chronic GVHD.
Collapse
Affiliation(s)
- Narendranath Epperla
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Hematology, The Ohio State University Medical Center, Columbus, Ohio
| | - Kwang Woo Ahn
- Center for International Blood and Marrow Transplant Research, Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Philippe Armand
- Department of Medical Oncology/Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Samantha Jaglowski
- Division of Hematology, The Ohio State University Medical Center, Columbus, Ohio
| | - Sairah Ahmed
- Department of Stem Cell Transplantation, MD Anderson Cancer Center, Houston, Texas
| | - Vaishalee P Kenkre
- Division of Hematology and Oncology, University of Wisconsin, Madison, Wisconsin
| | - Bipin Savani
- Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Madan Jagasia
- Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nirav N Shah
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Timothy S Fenske
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anna Sureda
- Servei d'Hematologica, Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain
| | - Sonali M Smith
- Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois
| | - Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
60
|
Schroeder MA, Choi J, Staser K, DiPersio JF. The Role of Janus Kinase Signaling in Graft-Versus-Host Disease and Graft Versus Leukemia. Biol Blood Marrow Transplant 2017; 24:1125-1134. [PMID: 29289756 DOI: 10.1016/j.bbmt.2017.12.797] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/20/2017] [Indexed: 12/18/2022]
Abstract
For patients with hematologic malignancies, allogeneic hematopoietic cell transplantation (alloHCT) offers a potential curative treatment option, primarily due to an allogeneic immune response against recipient tumor cells (ie, graft-versus-leukemia [GVL] activity). However, many recipients of alloHCT develop graft-versus-host disease (GVHD), in which allogeneic immune responses lead to the damage of healthy tissue. GVHD is a leading cause of nonrelapse mortality and a key contributor to morbidity among patients undergoing alloHCT. Therefore, improving alloHCT outcomes will require treatment strategies that prevent or mitigate GVHD without disrupting GVL activity. Janus kinases (JAKs) are intracellular signaling molecules that are well positioned to regulate GVHD. A variety of cytokines that signal through the JAK signaling pathways play a role in regulating the development, proliferation, and activation of several immune cell types important for GVHD pathogenesis, including dendritic cells, macrophages, T cells, B cells, and neutrophils. Importantly, despite JAK regulation of GVHD, preclinical evidence suggests that JAK inhibition preserves GVL activity. Here we provide an overview of potential roles for JAK signaling in the pathogenesis of acute and chronic GVHD as well as effects on GVL activity. We also review preclinical and clinical results with JAK inhibitors in acute and chronic GVHD settings, with added focus on those actively being evaluated in patients with acute and chronic GVHD.
Collapse
Affiliation(s)
- Mark A Schroeder
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri.
| | - Jaebok Choi
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri
| | - Karl Staser
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri
| | - John F DiPersio
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri
| |
Collapse
|
61
|
Affiliation(s)
- Robert Zeiser
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| | - Bruce R Blazar
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| |
Collapse
|
62
|
Bikel S, Jacobo-Albavera L, Sánchez-Muñoz F, Cornejo-Granados F, Canizales-Quinteros S, Soberón X, Sotelo-Mundo RR, Del Río-Navarro BE, Mendoza-Vargas A, Sánchez F, Ochoa-Leyva A. A novel approach for human whole transcriptome analysis based on absolute gene expression of microarray data. PeerJ 2017; 5:e4133. [PMID: 29230367 PMCID: PMC5724404 DOI: 10.7717/peerj.4133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/14/2017] [Indexed: 12/13/2022] Open
Abstract
Background In spite of the emergence of RNA sequencing (RNA-seq), microarrays remain in widespread use for gene expression analysis in the clinic. There are over 767,000 RNA microarrays from human samples in public repositories, which are an invaluable resource for biomedical research and personalized medicine. The absolute gene expression analysis allows the transcriptome profiling of all expressed genes under a specific biological condition without the need of a reference sample. However, the background fluorescence represents a challenge to determine the absolute gene expression in microarrays. Given that the Y chromosome is absent in female subjects, we used it as a new approach for absolute gene expression analysis in which the fluorescence of the Y chromosome genes of female subjects was used as the background fluorescence for all the probes in the microarray. This fluorescence was used to establish an absolute gene expression threshold, allowing the differentiation between expressed and non-expressed genes in microarrays. Methods We extracted the RNA from 16 children leukocyte samples (nine males and seven females, ages 6-10 years). An Affymetrix Gene Chip Human Gene 1.0 ST Array was carried out for each sample and the fluorescence of 124 genes of the Y chromosome was used to calculate the absolute gene expression threshold. After that, several expressed and non-expressed genes according to our absolute gene expression threshold were compared against the expression obtained using real-time quantitative polymerase chain reaction (RT-qPCR). Results From the 124 genes of the Y chromosome, three genes (DDX3Y, TXLNG2P and EIF1AY) that displayed significant differences between sexes were used to calculate the absolute gene expression threshold. Using this threshold, we selected 13 expressed and non-expressed genes and confirmed their expression level by RT-qPCR. Then, we selected the top 5% most expressed genes and found that several KEGG pathways were significantly enriched. Interestingly, these pathways were related to the typical functions of leukocytes cells, such as antigen processing and presentation and natural killer cell mediated cytotoxicity. We also applied this method to obtain the absolute gene expression threshold in already published microarray data of liver cells, where the top 5% expressed genes showed an enrichment of typical KEGG pathways for liver cells. Our results suggest that the three selected genes of the Y chromosome can be used to calculate an absolute gene expression threshold, allowing a transcriptome profiling of microarray data without the need of an additional reference experiment. Discussion Our approach based on the establishment of a threshold for absolute gene expression analysis will allow a new way to analyze thousands of microarrays from public databases. This allows the study of different human diseases without the need of having additional samples for relative expression experiments.
Collapse
Affiliation(s)
- Shirley Bikel
- Departamento de Microbiología Molecular, Universidad Nacional Autónoma de México, Instituto de Biotecnología, Cuernavaca, Morelos, México
| | - Leonor Jacobo-Albavera
- Instituto Nacional de Medicina Genómica, Instituto Nacional de Medicina Genómica, México City, México
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez (INCICh), México City, México
| | - Fernanda Cornejo-Granados
- Departamento de Microbiología Molecular, Universidad Nacional Autónoma de México, Instituto de Biotecnología, Cuernavaca, Morelos, México
| | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada la Salud, Instituto Nacional de Medicina Genómica, México City, México
| | - Xavier Soberón
- Instituto Nacional de Medicina Genómica, Instituto Nacional de Medicina Genómica, México City, México.,Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Rogerio R Sotelo-Mundo
- Laboratorio de Estructura Biomolecular, Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Hermosillo, Sonora, México
| | | | - Alfredo Mendoza-Vargas
- Instituto Nacional de Medicina Genómica, Instituto Nacional de Medicina Genómica, México City, México
| | - Filiberto Sánchez
- Departamento de Microbiología Molecular, Universidad Nacional Autónoma de México, Instituto de Biotecnología, Cuernavaca, Morelos, México
| | - Adrian Ochoa-Leyva
- Departamento de Microbiología Molecular, Universidad Nacional Autónoma de México, Instituto de Biotecnología, Cuernavaca, Morelos, México
| |
Collapse
|
63
|
Miklos D, Cutler CS, Arora M, Waller EK, Jagasia M, Pusic I, Flowers ME, Logan AC, Nakamura R, Blazar BR, Li Y, Chang S, Lal I, Dubovsky J, James DF, Styles L, Jaglowski S. Ibrutinib for chronic graft-versus-host disease after failure of prior therapy. Blood 2017; 130:2243-2250. [PMID: 28924018 PMCID: PMC6033048 DOI: 10.1182/blood-2017-07-793786] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/06/2017] [Indexed: 12/25/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a serious complication of allogeneic stem cell transplantation with few effective options available after failure of corticosteroids. B and T cells play a role in the pathophysiology of cGVHD. Ibrutinib inhibits Bruton tyrosine kinase in B cells and interleukin-2-inducible T-cell kinase in T cells. In preclinical models, ibrutinib reduced severity of cGVHD. This multicenter, open-label study evaluated the safety and efficacy of ibrutinib in patients with active cGVHD with inadequate response to corticosteroid-containing therapies. Forty-two patients who had failed 1 to 3 prior treatments received ibrutinib (420 mg) daily until cGVHD progression. The primary efficacy end point was cGVHD response based on 2005 National Institutes of Health criteria. At a median follow-up of 13.9 months, best overall response was 67%; 71% of responders showed a sustained response for ≥20 weeks. Responses were observed across involved organs evaluated. Most patients with multiple cGVHD organ involvement had a multiorgan response. Median corticosteroid dose in responders decreased from 0.29 mg/kg per day at baseline to 0.12 mg/kg per day at week 49; 5 responders discontinued corticosteroids. The most common adverse events were fatigue, diarrhea, muscle spasms, nausea, and bruising. Plasma levels of soluble factors associated with inflammation, fibrosis, and cGVHD significantly decreased over time with ibrutinib. Ibrutinib resulted in clinically meaningful responses with acceptable safety in patients with ≥1 prior treatments for cGVHD. Based on these results, ibrutinib was approved in the United States for treatment of adult patients with cGVHD after failure of 1 or more lines of systemic therapy. This trial was registered at www.clinicaltrials.gov as #NCT02195869.
Collapse
Affiliation(s)
- David Miklos
- Stanford University School of Medicine, Stanford, CA
| | | | - Mukta Arora
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | | | - Iskra Pusic
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | | | - Aaron C Logan
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | | | - Bruce R Blazar
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Yunfeng Li
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, CA; and
| | - Stephen Chang
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, CA; and
| | - Indu Lal
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, CA; and
| | - Jason Dubovsky
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, CA; and
| | | | - Lori Styles
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, CA; and
| | - Samantha Jaglowski
- Division of Hematology, The Ohio State University Cancer Center, Columbus, OH
| |
Collapse
|
64
|
Antibodies targeting surface membrane antigens in patients with chronic graft-versus-host disease. Blood 2017; 130:2889-2899. [PMID: 29138220 DOI: 10.1182/blood-2017-08-801001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) after allogeneic hematopoietic stem cell transplant reflects a complex immune response resulting in chronic damage to multiple tissues. Previous studies indicated that donor B cells and the antibodies they produce play an important role in the development of cGVHD. To understand the pathogenic role of antibodies in cGVHD, we focused our studies on posttransplant production of immunoglobulin G antibodies targeting cell surface antigens expressed in multiple cGVHD affected tissues, due to their potential functional impact on living cells in vivo. Using plate-bound cell membrane proteins as targets, we detected a significantly higher level of antibodies reactive with these membrane antigens in patients who developed cGVHD, compared with those who did not and healthy donors. Plasma-reactive antibody levels increased significantly prior to the clinical diagnosis of cGVHD and were reduced following cGVHD therapies including prednisone, interleukin-2, or extracorporeal photophoresis. Using cell-based immunoprecipitation with plasma from cGVHD patients and mass spectrometry, we identified 43 membrane proteins targeted by these antibodies. The presence of antibodies in cGVHD patients' plasma that specifically target 6 of these proteins was validated. Antibodies reactive with these 6 antigens were more frequently detected in patients with cGVHD compared with patients without cGVHD and healthy donors. These results indicate that antibodies that target membrane antigens of living cells frequently develop in cGVHD patients and further support a role for B cells and antibodies in the development of cGVHD.
Collapse
|
65
|
The Role of B Cell Targeting in Chronic Graft-Versus-Host Disease. Biomedicines 2017; 5:biomedicines5040061. [PMID: 29039818 PMCID: PMC5744085 DOI: 10.3390/biomedicines5040061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/20/2017] [Accepted: 10/09/2017] [Indexed: 11/17/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a leading cause of late morbidity and mortality following allogeneic stem cell transplantation. Current therapies, including corticosteroids and calcineurin inhibitors, are only effective in roughly 50% of cases; therefore, new treatment strategies are under investigation. What was previously felt to be a T cell disease has more recently been shown to involve activation of both T and B cells, as well as a number of cytokines. With a better understanding of its pathophysiology have come more expansive preclinical and clinical trials, many focused on B cell signaling. This report briefly reviews our current understanding of cGVHD pathophysiology and reviews clinical and preclinical trials with B cell-targeted agents.
Collapse
|
66
|
Reindl-Schwaighofer R, Heinzel A, Signorini L, Thaunat O, Oberbauer R. Mechanisms underlying human genetic diversity: consequence for antigraft antibody responses. Transpl Int 2017; 31:239-250. [DOI: 10.1111/tri.13059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 07/28/2017] [Accepted: 08/30/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis; Department of Internal Medicine III; Medical University of Vienna; Vienna Austria
| | - Andreas Heinzel
- Division of Nephrology and Dialysis; Department of Internal Medicine III; Medical University of Vienna; Vienna Austria
| | - Lorenzo Signorini
- Renal and Dialysis Unit; Department of Medicine; University of Verona; Verona Italy
| | - Olivier Thaunat
- Hospices Civils de Lyon; Hôpital Edouard Herriot; Service de Transplantation; Néphrologie et Immunologie Clinique; INSERM U1111; Université Lyon-I; Lyon France
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis; Department of Internal Medicine III; Medical University of Vienna; Vienna Austria
| |
Collapse
|
67
|
Friedrich P, Guerra-García P, Stetson A, Duncan C, Lehmann L. Young Female Donors Do Not Increase the Risk of Graft-versus-Host Disease or Impact Overall Outcomes in Pediatric HLA-Matched Sibling Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2017; 24:96-102. [PMID: 28958895 DOI: 10.1016/j.bbmt.2017.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/20/2017] [Indexed: 01/29/2023]
Abstract
Optimal donor selection is critical in hematopoietic stem cell transplantation (HSCT). Donor-recipient sex mismatch, donor age, and female donor-donor parity are known to impact graft-versus-host disease (GVHD) and outcomes in adults. Minor histocompatibility antigens encoded by the human Y chromosome can result in specific antibody formation in some female donors, may increase in frequency with increasing donor age, and may be contributory to the increased incidence of GVHD. To better understand the role of donor age/sex and sex matching in HSCT outcomes, we conducted a retrospective study of pediatric patients receiving their first myeloablative sibling donor HSCT (n = 244) from 1998 to 2012. Observed rates of GVHD were low: 17% of patients surviving past engraftment (n = 243) developed grades II to IV acute GVHD (aGVHD) and 14% surviving ≥ 100 days (n = 229) developed chronic GVHD (cGVHD). On multivariate analysis the risk of aGVHD, cGVHD, and death increased with patient age as expected. Female donor sex and sex mismatch (female donor-male recipient) had no impact on the development of aGVHD. cGVHD was increased with female donors only if the donor was ≥12 years old. No cGVHD was observed among 109 patients aged < 10 years who received a 6/6 HLA-matched marrow HSCT, regardless of donor age or sex. Survival was mostly driven by diagnosis. Results suggest that in pediatric HSCT, young HLA-matched siblings are equivalently good donors regardless of sex or donor-recipient sex mismatch.
Collapse
Affiliation(s)
- Paola Friedrich
- Department of Oncology and Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Pilar Guerra-García
- Department of Pediatric Hematology and Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Alyssa Stetson
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Christine Duncan
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Leslie Lehmann
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts.
| |
Collapse
|
68
|
Effect of Rituximab on Pulmonary Function in Bronchiolitis Obliterans Syndrome due to Graft-Versus-Host-Disease. Lung 2017; 195:781-788. [DOI: 10.1007/s00408-017-0051-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/06/2017] [Indexed: 12/30/2022]
|
69
|
Rituximab-based first-line treatment of cGVHD after allogeneic SCT: results of a phase 2 study. Blood 2017; 130:2186-2195. [PMID: 28864814 DOI: 10.1182/blood-2017-05-786137] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/21/2017] [Indexed: 01/14/2023] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is the main cause of late nonrelapse mortality and morbidity after allogeneic stem cell transplantation (allo-SCT). To improve such patients' outcomes, we conducted a phase 2, prospective, multicenter trial to test the efficacy of the addition of rituximab to corticosteroids (CSs) and cyclosporine A (CsA) as first-line therapy for newly diagnosed cGVHD after allo-SCT. Twenty-four patients (median age, 47 years) with mild (n = 2), moderate (n = 7), or severe (n = 15) cGVHD were included. All patients received rituximab 375 mg/m2 weekly for 4 weeks, followed by a second course 1 month later for patients with partial response. Twenty of 24 patients (83%) were in response at 1 year. Furthermore, among 19 evaluable patients, 14 (74%) were off CSs. The estimated 1-year overall survival was 83%, and the 1-year cumulative incidence of nonrelapse mortality was 14%. One patient died of progressive multifocal leukoencephalopathy. Although PD-L1hi naive B cells were significantly decreased at diagnosis of cGVHD, they increased after anti-CD20 B-cell depletion. In contrast, activated ICOShi PD-1hi circulating T follicular helper (Tfh) cells decreased after rituximab treatment. Overall, the addition of rituximab to corticosteroid and CsA appeared to be safe and effective for first-line treatment of cGVHD. Furthermore, our data suggest that this efficacy may be in part related to an effect on PD-L1hi B cells and Tfh cells. This study was registered at www.clinicaltrials.gov as identifier NCT01135641.
Collapse
|
70
|
InanlooRahatloo K, Liang G, Vo D, Ebert A, Nguyen I, Nguyen PK. Sex-based differences in myocardial gene expression in recently deceased organ donors with no prior cardiovascular disease. PLoS One 2017; 12:e0183874. [PMID: 28850583 PMCID: PMC5574577 DOI: 10.1371/journal.pone.0183874] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/12/2017] [Indexed: 02/08/2023] Open
Abstract
Sex differences in the development of the normal heart and the prevalence of cardiomyopathies have been reported. The molecular basis of these differences remains unclear. Sex differences in the human heart might be related to patterns of gene expression. Recent studies have shown that sex specific differences in gene expression in tissues including the brain, kidney, skeletal muscle, and liver. Similar data is limited for the heart. Herein we address this issue by analyzing donor and post-mortem adult human heart samples originating from 46 control individuals to study whole-genome gene expression in the human left ventricle. Using data from the genotype tissue expression (GTEx) project, we compared the transcriptome expression profiles of male and female hearts. We found that genes located on sex chromosomes were the most abundant ones among the sexually dimorphic genes. The majority of differentially expressed autosomal genes were those involved in the regulation of inflammation, which has been found to be an important contributor to left ventricular remodeling. Specifically, genes on autosomal chromosomes encoding chemokines with inflammatory functions (e.g. CCL4, CX3CL1, TNFAIP3) and a gene that regulates adhesion of immune cells to the endothelium (e.g., VCAM1) were identified with sex-specific expression levels. This study underlines the relevance of sex as an important modifier of cardiac gene expression. These results have important implications in the understanding of the differences in the physiology of the male and female heart transcriptome and how they may lead to different sex specific difference in human cardiac health and its control.
Collapse
Affiliation(s)
- Kolsoum InanlooRahatloo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Genetics, Genetic Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Grace Liang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Davis Vo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Antje Ebert
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ivy Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Patricia K. Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California, United States of America
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University and Veterans Affairs Palo Alto, Palo Alto, California, United States of America
- * E-mail: ,
| |
Collapse
|
71
|
An aberrant NOTCH2-BCR signaling axis in B cells from patients with chronic GVHD. Blood 2017; 130:2131-2145. [PMID: 28851699 DOI: 10.1182/blood-2017-05-782466] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/24/2017] [Indexed: 12/16/2022] Open
Abstract
B-cell receptor (BCR)-activated B cells contribute to pathogenesis in chronic graft-versus-host disease (cGVHD), a condition manifested by both B-cell autoreactivity and immune deficiency. We hypothesized that constitutive BCR activation precluded functional B-cell maturation in cGVHD. To address this, we examined BCR-NOTCH2 synergy because NOTCH has been shown to increase BCR responsiveness in normal mouse B cells. We conducted ex vivo activation and signaling assays of 30 primary samples from hematopoietic stem cell transplantation patients with and without cGVHD. Consistent with a molecular link between pathways, we found that BCR-NOTCH activation significantly increased the proximal BCR adapter protein BLNK. BCR-NOTCH activation also enabled persistent NOTCH2 surface expression, suggesting a positive feedback loop. Specific NOTCH2 blockade eliminated NOTCH-BCR activation and significantly altered NOTCH downstream targets and B-cell maturation/effector molecules. Examination of the molecular underpinnings of this "NOTCH2-BCR axis" in cGVHD revealed imbalanced expression of the transcription factors IRF4 and IRF8, each critical to B-cell differentiation and fate. All-trans retinoic acid (ATRA) increased IRF4 expression, restored the IRF4-to-IRF8 ratio, abrogated BCR-NOTCH hyperactivation, and reduced NOTCH2 expression in cGVHD B cells without compromising viability. ATRA-treated cGVHD B cells had elevated TLR9 and PAX5, but not BLIMP1 (a gene-expression pattern associated with mature follicular B cells) and also attained increased cytosine guanine dinucleotide responsiveness. Together, we reveal a mechanistic link between NOTCH2 activation and robust BCR responses to otherwise suboptimal amounts of surrogate antigen. Our findings suggest that peripheral B cells in cGVHD patients can be pharmacologically directed from hyperactivation toward maturity.
Collapse
|
72
|
Næss Ø, Mortensen LH, Vikanes Å, Smith GD. Offspring sex and parental health and mortality. Sci Rep 2017; 7:5285. [PMID: 28706249 PMCID: PMC5509737 DOI: 10.1038/s41598-017-05161-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 05/25/2017] [Indexed: 01/07/2023] Open
Abstract
Increased mortality has been observed in mothers and fathers with male offspring but little is known regarding specific diseases. In a register linkage we linked women born 1925–1954 having survived to age 50 (n = 661,031) to offspring and fathers (n = 691,124). Three approaches were used: 1) number of total boy and girl offspring, 2) sex of the first and second offspring and 3) proportion of boys to total number of offspring. A sub-cohort (n = 50,736 mothers, n = 44,794 fathers) from survey data was analysed for risk factors. Mothers had increased risk of total and cardiovascular mortality that was consistent across approaches: cardiovascular mortality of 1.07 (95% CI: 1.03–1.11) per boy (approach 2), 1.04 (1.01–1.07) if the first offspring was a boy, and 1.06 (1.01–1.10) if the first two offspring were boys (approach 3). We found that sex of offspring was not associated with total or cardiovascular mortality in fathers. For other diseases or risk factors no robust associations were seen in mothers or fathers. Increased cardiovascular risk in mothers having male offspring suggests a maternal disease specific mechanism. The lack of consistent associations on measured risk factors could suggest other biological pathways than those studied play a role in generating this additional cardiovascular risk.
Collapse
Affiliation(s)
- Øyvind Næss
- Epidemiological Division, National Institute of Public Health, Oslo, Norway. .,Institute of Health and Society, University of Oslo, Oslo, Norway.
| | - Laust H Mortensen
- Department of Public Health, University of Copenhagen, København, Denmark
| | - Åse Vikanes
- Epidemiological Division, National Institute of Public Health, Oslo, Norway.,The Intervention Center, Oslo University Hospital, Oslo, Norway
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, School of Social and Community Medicine, Bristol, UK
| |
Collapse
|
73
|
MacDonald KP, Blazar BR, Hill GR. Cytokine mediators of chronic graft-versus-host disease. J Clin Invest 2017; 127:2452-2463. [PMID: 28665299 DOI: 10.1172/jci90593] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Substantial preclinical and clinical research into chronic graft-versus-host disease (cGVHD) has come to fruition in the last five years, generating a clear understanding of a complex cytokine-driven cellular network. cGVHD is mediated by naive T cells differentiating within IL-17-secreting T cell and follicular Th cell paradigms to generate IL-21 and IL-17A, which drive pathogenic germinal center (GC) B cell reactions and monocyte-macrophage differentiation, respectively. cGVHD pathogenesis includes thymic damage, impaired antigen presentation, and a failure in IL-2-dependent Treg homeostasis. Pathogenic GC B cell and macrophage reactions culminate in antibody formation and TGF-β secretion, respectively, leading to fibrosis. This new understanding permits the design of rational cytokine and intracellular signaling pathway-targeted therapeutics, reviewed herein.
Collapse
Affiliation(s)
- Kelli Pa MacDonald
- Antigen Presentation and Immunoregulation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Bruce R Blazar
- Masonic Cancer Center; and Division of Blood and Marrow Transplantation, Department of Pediatrics; University of Minnesota, Minneapolis, USA
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
74
|
Donor and recipient age, gender and ABO incompatibility regardless of donor source: validated criteria for donor selection for haematopoietic transplants. Leukemia 2017. [PMID: 28642591 DOI: 10.1038/leu.2017.199] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Prior data indicate similar outcomes after transplants from human leukocyte antigen (HLA)-haplotype-matched relatives, HLA-identical siblings and HLA-matched unrelated donors. We used our prospective data set to answer a clinically important question: who is the best donor for a person with acute leukaemia transplanted in first complete remission. Patients were randomly divided into training (n=611) and validation (n=588) sets. A total of 1199 consecutive subjects received a transplant from an HLA-haplotype-matched relative using granulocyte colony-stimulating factor and anti-thymocyte globulin (n=685) or an HLA-identical sibling (n=514); 3-year leukaemia-free survivals (LFSs) were 75 and 74% (P=0.95), respectively. The multivariate model identified three major risk factors for transplant-related mortality (TRM): older donor/recipient age, female-to-male transplants and donor-recipient ABO major-mismatch transplants. A risk score was developed based on these three features. TRMs were 8%, 15% and 31% for subjects with scores of 0-1, 2 and 3, respectively, (P<0.001). Three-year LFSs were 78%, 74% and 58%, respectively, (P=0.003). The risk score was validated in an independent cohort. In conclusion, our data confirm donor source is not significantly correlated with transplant outcomes. Selection of the best donor needs to consider donor-recipient age, matching for gender and ABO incompatibility among persons with acute leukaemia receiving related transplants under our transplant modality.
Collapse
|
75
|
Gea-Banacloche J, Komanduri KV, Carpenter P, Paczesny S, Sarantopoulos S, Young JA, El Kassar N, Le RQ, Schultz KR, Griffith LM, Savani BN, Wingard JR. National Institutes of Health Hematopoietic Cell Transplantation Late Effects Initiative: The Immune Dysregulation and Pathobiology Working Group Report. Biol Blood Marrow Transplant 2017; 23:870-881. [PMID: 27751936 PMCID: PMC5392182 DOI: 10.1016/j.bbmt.2016.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/05/2016] [Indexed: 12/11/2022]
Abstract
Immune reconstitution after hematopoietic stem cell transplantation (HCT) beyond 1 year is not completely understood. Many transplant recipients who are free of graft-versus-host disease (GVHD) and not receiving any immunosuppression more than 1 year after transplantation seem to be able to mount appropriate immune responses to common pathogens and respond adequately to immunizations. However, 2 large registry studies over the last 2 decades seem to indicate that infection is a significant cause of late mortality in some patients, even in the absence of concomitant GVHD. Research on this topic is particularly challenging for several reasons. First, there are not enough long-term follow-up clinics able to measure even basic immune parameters late after HCT. Second, the correlation between laboratory measurements of immune function and infections is not well known. Third, accurate documentation of infectious episodes is notoriously difficult. Finally, it is unclear what measures can be implemented to improve the immune response in a clinically relevant way. A combination of long-term multicenter prospective studies that collect detailed infectious data and store samples as well as a national or multinational registry of clinically significant infections (eg, vaccine-preventable severe infections, opportunistic infections) could begin to address our knowledge gaps. Obtaining samples for laboratory evaluation of the immune system should be both calendar and eventdriven. Attention to detail and standardization of practices regarding prophylaxis, diagnosis, and definitions of infections would be of paramount importance to obtain clean reliable data. Laboratory studies should specifically address the neogenesis, maturation, and exhaustion of the adaptive immune system and, in particular, how these are influenced by persistent alloreactivity, inflammation, and viral infection. Ideally, some of these long-term prospective studies would collect information on long-term changes in the gut microbiome and their influence on immunity. Regarding enhancement of immune function, prospective measurement of the response to vaccines late after HCT in a variety of clinical settings should be undertaken to better understand the benefits as well as the limitations of immunizations. The role of intravenous immunoglobulin is still not well defined, and studies to address it should be encouraged.
Collapse
Affiliation(s)
- Juan Gea-Banacloche
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, Maryland.
| | - Krishna V Komanduri
- Sylvester Adult Stem Cell Transplant Program, University of Miami, Coral Gables, Florida
| | - Paul Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; University of Washington School of Medicine Pediatrics, Seattle, Washington
| | - Sophie Paczesny
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Stefanie Sarantopoulos
- Division of Hematological Malignancies and Cellular Therapy, Duke University Department of Medicine and Duke Cancer Institute, Durham, North Carolina
| | - Jo-Anne Young
- Division of Infectious Diseases and International Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Nahed El Kassar
- National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Robert Q Le
- Medical Officer, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Kirk R Schultz
- Professor of Pediatrics, UBC, Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital and Research Institute, Vancouver, Canada
| | - Linda M Griffith
- Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bipin N Savani
- Long Term Transplant Clinic, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John R Wingard
- University of Florida Health Cancer Center, Gainesville, Florida; Bone Marrow Transplant Program, Division of Hematology/Oncology, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
76
|
Kolb HJ. Hematopoietic stem cell transplantation and cellular therapy. HLA 2017; 89:267-277. [DOI: 10.1111/tan.13005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 01/08/2023]
Affiliation(s)
- H.-J. Kolb
- Helmholtz Zentrum Muenchen; Muenchen Germany
- Ludwig Maximilians Universitaet Muenchen; Muenchen Germany
- Klinikum Muenchen Schwabing Muenchen; Muenchen Germany
- Department PediatricsTechnische Unoiversitaet Muenchen; Muenchen Germany
| |
Collapse
|
77
|
Ghimire S, Weber D, Mavin E, Wang XN, Dickinson AM, Holler E. Pathophysiology of GvHD and Other HSCT-Related Major Complications. Front Immunol 2017; 8:79. [PMID: 28373870 PMCID: PMC5357769 DOI: 10.3389/fimmu.2017.00079] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
For over 60 years, hematopoietic stem cell transplantation has been the major curative therapy for several hematological and genetic disorders, but its efficacy is limited by the secondary disease called graft versus host disease (GvHD). Huge advances have been made in successful transplantation in order to improve patient quality of life, and yet, complete success is hard to achieve. This review assimilates recent updates on pathophysiology of GvHD, prophylaxis and treatment of GvHD-related complications, and advances in the potential treatment of GvHD.
Collapse
Affiliation(s)
- Sakhila Ghimire
- Department of Internal Medicine III, University Medical Centre , Regensburg , Germany
| | - Daniela Weber
- Department of Internal Medicine III, University Medical Centre , Regensburg , Germany
| | - Emily Mavin
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle , UK
| | - Xiao Nong Wang
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle , UK
| | - Anne Mary Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle , UK
| | - Ernst Holler
- Department of Internal Medicine III, University Medical Centre , Regensburg , Germany
| |
Collapse
|
78
|
Pirfenidone ameliorates murine chronic GVHD through inhibition of macrophage infiltration and TGF-β production. Blood 2017; 129:2570-2580. [PMID: 28254742 DOI: 10.1182/blood-2017-01-758854] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 02/23/2017] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is hampered by chronic graft-versus-host disease (cGVHD), resulting in multiorgan fibrosis and diminished function. Fibrosis in lung and skin leads to progressive bronchiolitis obliterans (BO) and scleroderma, respectively, for which new treatments are needed. We evaluated pirfenidone, a Food and Drug Administration (FDA)-approved drug for idiopathic pulmonary fibrosis, for its therapeutic effect in cGVHD mouse models with distinct pathophysiology. In a full major histocompatibility complex (MHC)-mismatched, multiorgan system model with BO, donor T-cell responses that support pathogenic antibody production are required for cGVHD development. Pirfenidone treatment beginning one month post-transplant restored pulmonary function and reversed lung fibrosis, which was associated with reduced macrophage infiltration and transforming growth factor-β production. Pirfenidone dampened splenic germinal center B-cell and T-follicular helper cell frequencies that collaborate to produce antibody. In both a minor histocompatibility antigen-mismatched as well as a MHC-haploidentical model of sclerodermatous cGVHD, pirfenidone significantly reduced macrophages in the skin, although clinical improvement of scleroderma was only seen in one model. In vitro chemotaxis assays demonstrated that pirfenidone impaired macrophage migration to monocyte chemoattractant protein-1 (MCP-1) as well as IL-17A, which has been linked to cGVHD generation. Taken together, our data suggest that pirfenidone is a potential therapeutic agent to ameliorate fibrosis in cGVHD.
Collapse
|
79
|
da Silva MB, da Cunha FF, Terra FF, Camara NOS. Old game, new players: Linking classical theories to new trends in transplant immunology. World J Transplant 2017; 7:1-25. [PMID: 28280691 PMCID: PMC5324024 DOI: 10.5500/wjt.v7.i1.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/16/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
The evolutionary emergence of an efficient immune system has a fundamental role in our survival against pathogenic attacks. Nevertheless, this same protective mechanism may also establish a negative consequence in the setting of disorders such as autoimmunity and transplant rejection. In light of the latter, although research has long uncovered main concepts of allogeneic recognition, immune rejection is still the main obstacle to long-term graft survival. Therefore, in order to define effective therapies that prolong graft viability, it is essential that we understand the underlying mediators and mechanisms that participate in transplant rejection. This multifaceted process is characterized by diverse cellular and humoral participants with innate and adaptive functions that can determine the type of rejection or promote graft acceptance. Although a number of mediators of graft recognition have been described in traditional immunology, recent studies indicate that defining rigid roles for certain immune cells and factors may be more complicated than originally conceived. Current research has also targeted specific cells and drugs that regulate immune activation and induce tolerance. This review will give a broad view of the most recent understanding of the allogeneic inflammatory/tolerogenic response and current insights into cellular and drug therapies that modulate immune activation that may prove to be useful in the induction of tolerance in the clinical setting.
Collapse
|
80
|
Cooke KR, Luznik L, Sarantopoulos S, Hakim FT, Jagasia M, Fowler DH, van den Brink MRM, Hansen JA, Parkman R, Miklos DB, Martin PJ, Paczesny S, Vogelsang G, Pavletic S, Ritz J, Schultz KR, Blazar BR. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 23:211-234. [PMID: 27713092 PMCID: PMC6020045 DOI: 10.1016/j.bbmt.2016.09.023] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
Chronic graft-versus-host disease (GVHD) is the leading cause of late, nonrelapse mortality and disability in allogeneic hematopoietic cell transplantation recipients and a major obstacle to improving outcomes. The biology of chronic GVHD remains enigmatic, but understanding the underpinnings of the immunologic mechanisms responsible for the initiation and progression of disease is fundamental to developing effective prevention and treatment strategies. The goals of this task force review are as follows: This document is intended as a review of our understanding of chronic GVHD biology and therapies resulting from preclinical studies, and as a platform for developing innovative clinical strategies to prevent and treat chronic GVHD.
Collapse
Affiliation(s)
- Kenneth R Cooke
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland.
| | - Leo Luznik
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Stefanie Sarantopoulos
- Division of Hematological Malignancies and Cellular Therapy, Department of Immunology and Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Frances T Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Madan Jagasia
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Marcel R M van den Brink
- Departments of Immunology and Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John A Hansen
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Robertson Parkman
- Division of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford University, Palo Alto, California
| | - David B Miklos
- Division of Blood and Marrow Transplantation, Stanford University, Palo Alto, California
| | - Paul J Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Sophie Paczesny
- Departments of Pediatrics and Immunology, Wells Center for Pediatric Research, Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Georgia Vogelsang
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Steven Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kirk R Schultz
- Michael Cuccione Childhood Cancer Research Program, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
81
|
Grønningsæter IS, Tsykunova G, Lilleeng K, Ahmed AB, Bruserud Ø, Reikvam H. Bronchiolitis obliterans syndrome in adults after allogeneic stem cell transplantation-pathophysiology, diagnostics and treatment. Expert Rev Clin Immunol 2017; 13:553-569. [DOI: 10.1080/1744666x.2017.1279053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ida Sofie Grønningsæter
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Galina Tsykunova
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
| | - Kyrre Lilleeng
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
| | - Aymen Bushra Ahmed
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
| | - Øystein Bruserud
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | |
Collapse
|
82
|
Juric MK, Shevtsov M, Mozes P, Ogonek J, Crossland RE, Dickinson AM, Greinix HT, Holler E, Weissinger EM, Multhoff G. B-Cell-Based and Soluble Biomarkers in Body Liquids for Predicting Acute/Chronic Graft-versus-Host Disease after Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2017; 7:660. [PMID: 28138325 PMCID: PMC5238459 DOI: 10.3389/fimmu.2016.00660] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/16/2016] [Indexed: 02/02/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the main curative therapy for hematological malignancy such as leukemias, lymphomas, or multiple myelomas and some other hematological disorders. In this therapy, cure of hematological diseases relies on graft-versus-malignancy effects by allogenic immune cells. However, severe posttransplant treatment-associated complications such as acute graft-versus-host disease (aGvHD) and chronic graft-versus-host disease (cGvHD) limit this approach. Most research into GvHD has concentrated on the aGvHD, while the more complex and multifaceted chronic form has been largely poorly investigated. cGvHD is a multi-organ autoimmune disorder and is the major cause of non-relapse morbidity and mortality following allo-HSCT, occurring in about 50% of patients, or 13,000–15,000 patients per year worldwide. Therefore, there is a high medical need for an early prediction of these therapy-associated toxicities. Biomarkers have gained importance over the last decade in diagnosis, in prognosis, and in prediction of pending diseases or side effects. Biomarkers can be cells, factors isolated from target tissues, or soluble factors that can be detected in body fluids. In this review, we aim to summarize some of the recent developments of biomarkers in the field of allo-HSCT. We will focus on cell-based biomarkers (B-cell subsets) for cGvHD and soluble factors including microRNA (miRNA), which are excreted into serum/plasma and urine. We also discuss the potential role of cytosolic and extracellular 70 kDa heat shock proteins (HSP70) as potential biomarkers for aGvHD and their role in preclinical models. Proteomic biomarkers in the blood have been used as predictors of treatment responses in patients with aGvHD for many years. More recently, miRNAs have been found to serve as a biomarker to diagnose aGvHD in the plasma. Another development relates to urine-based biomarkers that are usually detected by capillary electrophoresis and mass spectrometry. These biomarkers have the potential to predict the development of severe aGvHD (grades III–IV), overall mortality, and the pending development of cGvHD in patients posttransplant.
Collapse
Affiliation(s)
- Mateja Kralj Juric
- Department of Internal Medicine I, BMT, Medical University of Vienna , Vienna , Austria
| | - Maxim Shevtsov
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Petra Mozes
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Justyna Ogonek
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Transplantation Biology, Hannover Medical School , Hannover , Germany
| | - Rachel E Crossland
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Anne M Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | | | - Ernst Holler
- Department of Internal Medicine III, University Hospital of Regensburg , Regensburg , Germany
| | - Eva M Weissinger
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Transplantation Biology, Hannover Medical School , Hannover , Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| |
Collapse
|
83
|
Atilla E, Ataca Atilla P, Demirer T. A Review of Myeloablative vs Reduced Intensity/Non-Myeloablative Regimens in Allogeneic Hematopoietic Stem Cell Transplantations. Balkan Med J 2017; 34:1-9. [PMID: 28251017 PMCID: PMC5322516 DOI: 10.4274/balkanmedj.2017.0055] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) is a curative treatment option for both malignant and some benign hematological diseases. During the last decade, many of the newer high-dose regimens in different intensity have been developed specifically for patients with hematologic malignancies and solid tumors. Today there are three main approaches used prior to allogeneic transplantation: Myeloablative (MA), Reduced Intensity Conditioning (RIC) and Non-MA (NMA) regimens. MA regimens cause irreversible cytopenia and there is a requirement for stem cell support. Patients who receive NMA regimen have minimal cytopenia and this type of regimen can be given without stem cell support. RIC regimens do not fit the criteria of MA and NMA: the cytopenia is reversible and the stem cell support is necessary. NMA/RIC for Allo-HSCT has opened a new era for treating elderly patients and those with comorbidities. The RIC conditioning was used for 40% of all Allo-HSCT and this trend continue to increase. In this paper, we will review these regimens in the setting of especially allogeneic HSCT and our aim is to describe the history, features and impact of these conditioning regimens on specific diseases.
Collapse
Affiliation(s)
- Erden Atilla
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Pınar Ataca Atilla
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Taner Demirer
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
84
|
Assandri R, Serana F, Montanelli A. Development of PBC/SSc overlap syndrome in chronic GVHD patient: immunological implications in the presence of mitochondrial, nucleolar and spindle midzone autoantigens. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2017; 10:323-331. [PMID: 29379599 PMCID: PMC5758742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Chronic Graft versus Host Disease (cGVHD) is a complex disease resulting from donor T-cell recognition of a genetically disparate recipient that is unable to reject donor cells after allogeneic Stem Cell Transplantation (HSCT). cGVHD has some features resembling to autoimmune diseases (AD) such as Sjögren syndrome, primary biliary cirrhosis (PBC) and scleroderma (SSc). Also patients with cGVHD could develop extensive cGVHD with scleroderma-like skin manifestations and other clinical signs similar to those of patients with scleroderma. We take into consideration a patient with GVHD that developed PBC/SSc overlap syndrome with a complex and particular autoantibodies profile. Indirect immunofluorescence (IIF) with double coloration showed a cytoplasmic mitochondrial-like pattern, a clumpy nucleolar staining pattern, and a cell-cycle related staining pattern. Following anaphase onset, proteins regulator of cytokinesis localizes to the overlap zone on the ends of midzone microtubules and becomes compacted during furrow ingression to form the midbody. Second level tests confirmed the presence of anti-mitochondrial antibodies M2-subunit but no other autoantibodies were found. We performed a home-made immunoblot analysis that identified a 37 kDa fibrillarin band, and not identify 47 kDa, 31KDa and 18/20 kDa bands. After literature review of these possible cellular localizations, the proteins recognized by our patient's serum seem likely to be Aab to core midzone organizer components. However, due to the unavailability of the proper techniques in our laboratory, we were not able to further characterize them. The pathogenesis and morbidity of cGVHD after HSCT remains enigmatic, but the presence of specific autoantibodies are the hallmark of AD and represent a possibility of differential diagnosis. Standard techniques combined with the use of non-routinely laboratory techniques are a usefully and complementary method for studying difficult and particular cases. In fact, these autoantibodies will be considered as ''diagnostic'' and not as ''esoteric'' antibodies. In conclusion, a re-assessment of the diagnostic protocols in cGVHD together with a precise observation of the clinical and laboratory picture will ultimately help us clarify the disease and could provide a better understanding of the immune network deregulation.
Collapse
Affiliation(s)
- Roberto Assandri
- Clinical Investigation Laboratory, ASST Crema, Crema, Cremona, Italy
| | - Federico Serana
- Clinical Investigation Laboratory, Diagnostics Department, Spedali Civili of Brescia, Brescia, Italy
| | - Alessandro Montanelli
- Clinical Investigation Laboratory, Diagnostics Department, Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
85
|
Belle L, Fransolet G, Somja J, Binsfeld M, Delvenne P, Drion P, Hannon M, Beguin Y, Ehx G, Baron F. Limited Impact of Imatinib in a Murine Model of Sclerodermatous Chronic Graft-versus-Host Disease. PLoS One 2016; 11:e0167997. [PMID: 27942010 PMCID: PMC5152855 DOI: 10.1371/journal.pone.0167997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 11/23/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Sclerodermatous chronic Graft-versus-Host Disease (scl-cGVHD) is one of the most severe form of cGVHD. The Platelet-derived Grotwth Factor (PDGF) and the Transforming Growth Factor-β (TGF-β) play a significant role in the fibrosing process occurring in scl-cGVHD. This prompted us to assess the impact of the PDGF-r and c-Abl tyrosine kinase inhibitor imatinib on scl-cGVHD. METHODS To assess the impact of imatinib on T cell subset proliferation in vivo, Balb/cJ recipient mice were lethally (7 Gy) irradiated and then injected with 10x106 bone marrow cells from B10.D2 mice on day 0. Fourteen days later, 70x106 carboxyfluorescein succinimidyl ester (CFSE)-labeled splenocytes from B10.D2 mice were infused and imatinib or sterile water was administered for 5 days. To induce severe scl-cGVHD, Balb/cJ mice were injected i.v. with 10.106 bone marrow cells and 70.106 splenocytes from B10.D2 donor mice after 7 Gy irradiation. Mice were then given sterile water or imatinib from day +7 after transplantation to the end of the experiment (day +52). RESULTS Imatinib decreased the proliferation of total T cells (P = 0.02), CD8+ T cells (P = 0.01), and of regulatory T cells (Tregs) (P = 0.02) in the spleen. In the severe scl-cGVHD model, imatinib-treated mice had significantly lower levels of PDGF-r phosphorylation than control mice on day 29 after transplantation (P = 0.008). However, scl-cGVHD scores were similar between vehicle- and imatinib-treated mice during the whole experiment, while there was a suggestion for less weight loss in imatinib-treated mice that reached statistical significance at day +52 following transplantation (P = 0.02). CONCLUSIONS Imatinib had a limited impact in murine scl-cGVHD despite significant inhibition of PDGF-r.
Collapse
Affiliation(s)
- Ludovic Belle
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Gilles Fransolet
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- * E-mail:
| | - Joan Somja
- Department of Pathology, University of Liège, Liège, Belgium
| | - Marilène Binsfeld
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | | | | | - Muriel Hannon
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Yves Beguin
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- Department of Medicine, Division of Hematology, CHU of Liège, Liège, Belgium
| | - Grégory Ehx
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Frédéric Baron
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- Department of Medicine, Division of Hematology, CHU of Liège, Liège, Belgium
| |
Collapse
|
86
|
Bohmann EM, Fehn U, Holler B, Weber D, Holler E, Herr W, Hoffmann P, Edinger M, Wolff D. Altered immune reconstitution of B and T cells precedes the onset of clinical symptoms of chronic graft-versus-host disease and is influenced by the type of onset. Ann Hematol 2016; 96:299-310. [PMID: 27942862 DOI: 10.1007/s00277-016-2881-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 11/14/2016] [Indexed: 10/20/2022]
Abstract
We analyzed lymphocyte subpopulations and cytokines 3 months after allogeneic hematopoietic stem cell transplantation aiming to identify predictive cellular and serum markers for chronic graft-versus-host disease (cGVHD). Samples of 49 patients (pts) (no cGVHD (n = 14), subsequent quiescent onset (n = 16), de novo onset of cGVHD (n = 19)) were analyzed in the absence of active GVHD by flow cytometry and enzyme-linked immunosorbent assay. All mean absolute cell counts are presented as cells per microliter; relative cell counts are presented as percentage of lymphocytes. Pts with subsequent de novo cGVHD had significantly higher relative and absolute counts of CD4+ T cells including higher absolute counts of CD4+ memory T cells (22.36%; 206.55/μl; 136/μl, respectively) compared to pts with subsequent quiescent onset of cGVHD (12.41%; 83.42/μl; 54.3/μl) and pts without cGVHD (10.55%) with regard to relative counts of CD4+ T cells. Similarly, significantly more relative and absolute regulatory T cell numbers (CD4+FOXP3+) were detected in pts with de novo onset of cGVHD (3.08% and 24.63/μl) compared to those in pts without (1.25% and 9.06/μl) or with quiescent onset of cGVHD (1.15% and 6.91/μl). Finally, relative B cell counts, including naïve and memory B cells, were also significantly decreased in pts developing quiescent cGVHD (0.85, 0.73, 0.12% resp.) when compared to pts with de novo onset (5.61, 5.24, 0.38%). The results demonstrate that alterations in immune reconstitution are already present before onset of clinical symptoms and differ between de novo and quiescent onset of disease.
Collapse
Affiliation(s)
- E-M Bohmann
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany
| | - U Fehn
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany.,Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - B Holler
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany
| | - D Weber
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany
| | - E Holler
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany.,Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - W Herr
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany
| | - P Hoffmann
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany.,Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - M Edinger
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany.,Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - D Wolff
- Department of Internal Medicine III, University Hospital of Regensburg, F.J. Strauss Allee 11, 93053, Regensburg, Germany. .,Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
87
|
Im A, Hakim FT, Pavletic SZ. Novel targets in the treatment of chronic graft-versus-host disease. Leukemia 2016; 31:543-554. [PMID: 27899803 DOI: 10.1038/leu.2016.367] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022]
Abstract
Despite advances that have improved survival after allogeneic hematopoietic stem cell transplantation (HCT), chronic graft-versus-host disease (GVHD) remains a leading cause of late morbidity and mortality after transplant. Current treatment options show limited efficacy in steroid-refractory disease, and there exists a paucity of robust data to guide management decisions. Lack of United States Food and Drug Administration (FDA)- or European Medicines Agency (EMA)-approved agents in GVHD underscore the importance of developing novel therapies. Better understanding of the biology of chronic GVHD has provided novel targets for treatment, and structured guidelines in diagnosis and in clinical trial design have provided a common language and pathways for research in this area. These, combined with the surge of drug development in Oncology and Immunology, are factors that have contributed to the accelerating field of drug development and clinical research in chronic GVHD. In these exciting times, it is possible to foresee long awaited advances in the treatment of this devastating complication of HCT. This review will summarize the ongoing clinical development for novel therapies in chronic GVHD.
Collapse
Affiliation(s)
- A Im
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh Cancer Institute and UPMC Cancer Centers, Pittsburgh, PA, USA.,Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| | - F T Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| | - S Z Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
88
|
Juric MK, Ghimire S, Ogonek J, Weissinger EM, Holler E, van Rood JJ, Oudshoorn M, Dickinson A, Greinix HT. Milestones of Hematopoietic Stem Cell Transplantation - From First Human Studies to Current Developments. Front Immunol 2016; 7:470. [PMID: 27881982 PMCID: PMC5101209 DOI: 10.3389/fimmu.2016.00470] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
Since the early beginnings, in the 1950s, hematopoietic stem cell transplantation (HSCT) has become an established curative treatment for an increasing number of patients with life-threatening hematological, oncological, hereditary, and immunological diseases. This has become possible due to worldwide efforts of preclinical and clinical research focusing on issues of transplant immunology, reduction of transplant-associated morbidity, and mortality and efficient malignant disease eradication. The latter has been accomplished by potent graft-versus-leukemia (GvL) effector cells contained in the stem cell graft. Exciting insights into the genetics of the human leukocyte antigen (HLA) system allowed improved donor selection, including HLA-identical related and unrelated donors. Besides bone marrow, other stem cell sources like granulocyte-colony stimulating-mobilized peripheral blood stem cells and cord blood stem cells have been established in clinical routine. Use of reduced-intensity or non-myeloablative conditioning regimens has been associated with a marked reduction of non-hematological toxicities and eventually, non-relapse mortality allowing older patients and individuals with comorbidities to undergo allogeneic HSCT and to benefit from GvL or antitumor effects. Whereas in the early years, malignant disease eradication by high-dose chemotherapy or radiotherapy was the ultimate goal; nowadays, allogeneic HSCT has been recognized as cellular immunotherapy relying prominently on immune mechanisms and to a lesser extent on non-specific direct cellular toxicity. This chapter will summarize the key milestones of HSCT and introduce current developments.
Collapse
Affiliation(s)
- Mateja Kralj Juric
- BMT, Department of Internal Medicine I, Medical University of Vienna , Vienna , Austria
| | - Sakhila Ghimire
- Department of Internal Medicine III, University Hospital of Regensburg , Regensburg , Germany
| | - Justyna Ogonek
- Transplantation Biology, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School , Hannover , Germany
| | - Eva M Weissinger
- Transplantation Biology, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School , Hannover , Germany
| | - Ernst Holler
- Department of Internal Medicine III, University Hospital of Regensburg , Regensburg , Germany
| | - Jon J van Rood
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden , Netherlands
| | - Machteld Oudshoorn
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden , Netherlands
| | - Anne Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | | |
Collapse
|
89
|
Chronic graft-versus-host disease: biological insights from preclinical and clinical studies. Blood 2016; 129:13-21. [PMID: 27821504 DOI: 10.1182/blood-2016-06-686618] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/06/2016] [Indexed: 12/15/2022] Open
Abstract
With the increasing use of mismatched, unrelated, and granulocyte colony-stimulating factor-mobilized peripheral blood stem cell donor grafts and successful treatment of older recipients, chronic graft-versus-host disease (cGVHD) has emerged as the major cause of nonrelapse mortality and morbidity. cGVHD is characterized by lichenoid changes and fibrosis that affects a multitude of tissues, compromising organ function. Beyond steroids, effective treatment options are limited. Thus, new strategies to both prevent and treat disease are urgently required. Over the last 5 years, our understanding of cGVHD pathogenesis and basic biology, born out of a combination of mouse models and correlative clinical studies, has radically improved. We now understand that cGVHD is initiated by naive T cells, differentiating predominantly within highly inflammatory T-helper 17/T-cytotoxic 17 and T-follicular helper paradigms with consequent thymic damage and impaired donor antigen presentation in the periphery. This leads to aberrant T- and B-cell activation and differentiation, which cooperate to generate antibody-secreting cells that cause the deposition of antibodies to polymorphic recipient antigens (ie, alloantibody) or nonpolymorphic antigens common to both recipient and donor (ie, autoantibody). It is now clear that alloantibody can, in concert with colony-stimulating factor 1 (CSF-1)-dependent donor macrophages, induce a transforming growth factor β-high environment locally within target tissue that results in scleroderma and bronchiolitis obliterans, diagnostic features of cGVHD. These findings have yielded a raft of potential new therapeutics, centered on naive T-cell depletion, interleukin-17/21 inhibition, kinase inhibition, regulatory T-cell restoration, and CSF-1 inhibition. This new understanding of cGVHD finally gives hope that effective therapies are imminent for this devastating transplant complication.
Collapse
|
90
|
Ibrutinib efficacy and tolerability in patients with relapsed chronic lymphocytic leukemia following allogeneic HCT. Blood 2016; 128:2899-2908. [PMID: 27802969 DOI: 10.1182/blood-2016-06-715284] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/15/2016] [Indexed: 02/07/2023] Open
Abstract
Ibrutinib, a potent and irreversible small-molecule inhibitor of both Bruton's tyrosine kinase and interleukin-2 inducible kinase (ITK), has been used to treat relapsed/refractory chronic lymphocytic leukemia (CLL) with prolongation of progression-free and overall survival. Here, we present 27 patients with relapsed CLL following allogeneic hematopoietic cell transplant (HCT) who subsequently received ibrutinib salvage therapy. Sixteen of these patients were part of multi-institutional clinical trials and achieved an overall response rate of 87.5%. An additional 11 patients were treated at Stanford University following US Food and Drug Administration approval of ibrutinib; 7 (64%) achieved a complete response, and 3 (27%) achieved a partial response. Of the 9 patients treated at Stanford who had mixed chimerism-associated CLL relapse, 4 (44%) converted to full donor chimerism following ibrutinib initiation, in association with disease response. Four of 11 (36%) patients evaluated by ClonoSeq achieved minimal residual disease negativity with CLL <1/10 000 white blood cells, which persisted even after ibrutinib was discontinued, in 1 case even after 26 months. None of the 27 patients developed graft-versus-host-disease (GVHD) following ibrutinib initiation. We postulate that ibrutinib augments the graft-versus-leukemia (GVL) benefit through a T-cell-mediated effect, most likely due to ITK inhibition. To investigate the immune modulatory effects of ibrutinib, we completed comprehensive immune phenotype characterization of peripheral B and T cells from treated patients. Our results show that ibrutinib selectively targets pre-germinal B cells and depletes Th2 helper cells. Furthermore, these effects persisted after drug discontinuation. In total, our results provide evidence that ibrutinib effectively augments GVL without causing GVHD.
Collapse
|
91
|
Goryainov VA, Kaabak MM, Babenko NN, Morozova MM, Platova EN, Agureeva LI, Dymova OV, Panin VV. [The effect of gender on the results of related kidney transplantation]. Khirurgiia (Mosk) 2016:62-67. [PMID: 27296125 DOI: 10.17116/hirurgia2016662-67] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIM To define the effect of donor and recipient gender on the results of kidney transplantation from living related donor. MATERIAL AND METHODS Group of 271 patients who underwent kidney transplantation from living related donor was analyzed. There were 115 women and 156 men. Age varied from 1 to 63 years (mean 21.30±12.32). There were 127 children aged 1-18 years (mean 11.28±4.63) and 144 adults aged 19-63 years (mean 29.81±11.24). Donors included 162 women and 109 men. Overall survival was calculated using Kaplan-Mayer. Mortality and incidence of transplants failure were determined using Fisher's exact test. RESULTS All patients were divided into 2 groups depending on recipients' gender and then into 4 subgroups depending on gender of donors and recipients. Comparative statistical analysis showed that transplants survival was higher in women vs. men (T=2.7, p=0.007). Survival of patients was similar in both groups. Moreover it was the best in subgroup of recipients-women with kidneys from donors-men. Difference was statistically significant (T=2.16, p=0.03). There was no significant difference in all other cases. CONCLUSION The results of kidney transplantation are better in recipients-women than in men.
Collapse
Affiliation(s)
- V A Goryainov
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - M M Kaabak
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - N N Babenko
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - M M Morozova
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - E N Platova
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - L I Agureeva
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - O V Dymova
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - V V Panin
- B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| |
Collapse
|
92
|
Therapeutic regulatory T-cell adoptive transfer ameliorates established murine chronic GVHD in a CXCR5-dependent manner. Blood 2016; 128:1013-7. [PMID: 27385791 DOI: 10.1182/blood-2016-05-715896] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/30/2016] [Indexed: 12/15/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a major complication of allogeneic hematopoietic stem cell transplantation. In cGVHD, alloreactive T cells and germinal center (GC) B cells often participate in GC reactions to produce pathogenic antibodies. Although regulatory T cells (Tregs) can inhibit GC reactions, Treg numbers are reduced in cGVHD, contributing to cGVHD pathogenesis. Here, we explored 2 means to increase Tregs in cGVHD: interleukin-2/monoclonal antibody (IL-2/mAb) complexes and donor Treg infusions. IL-2/mAb complexes given over 1 month were efficacious in expanding Tregs and treating established cGVHD in a multi-organ-system disease mouse model characterized by GC reactions, antibody deposition, and lung dysfunction. In an acute GVHD (aGVHD) model, IL-2/mAb complexes given for only 4 days resulted in rapid mortality, indicating IL-2/mAb complexes can drive conventional T-cell (Tcon)-mediated injury. In contrast, Treg infusions, which uniformly suppress aGVHD, increased Treg frequency and were effective in preventing the onset of, and treating, established cGVHD. Efficacy was dependent upon CXCR5-sufficient Tregs homing to, and inhibiting, GC reactions. These studies indicate that the infusion of Tregs, especially ones enriched for GC homing, may be desirable for cGVHD therapy. Although IL-2/mAb complexes can be efficacious in cGVHD, a cautious approach needs to be taken in settings in which aGVHD elements, and associated Tcon, are present.
Collapse
|
93
|
Kennedy VE, Savani BN, Greer JP, Kassim AA, Engelhardt BG, Goodman SA, Sengsayadeth S, Chinratanalab W, Jagasia M. Reduced-Intensity Conditioning with Fludarabine, Cyclophosphamide, and Rituximab Is Associated with Improved Outcomes Compared with Fludarabine and Busulfan after Allogeneic Stem Cell Transplantation for B Cell Malignancies. Biol Blood Marrow Transplant 2016; 22:1801-1807. [PMID: 27377900 DOI: 10.1016/j.bbmt.2016.06.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/29/2016] [Indexed: 12/17/2022]
Abstract
Reduced-intensity conditioning (RIC) has been used increasingly for allogeneic hematopoietic cell transplantation to minimize transplant-related mortality while maintaining the graft-versus-tumor effect. In B cell lymphoid malignancies, reduced-intensity regimens containing rituximab, an antiCD20 antibody, have been associated with favorable survival; however, the long-term outcomes of rituximab-containing versus nonrituximab-containing regimens for allogeneic hematopoietic cell transplantation in B cell lymphoid malignancies remain to be determined. We retrospectively analyzed 94 patients who received an allogeneic transplant for a B cell lymphoid malignancy. Of these, 33 received RIC with fludarabine, cyclophosphamide, and rituximab (FCR) and graft-versus-host disease (GVHD) prophylaxis with a calcineurin inhibitor and mini-methotrexate, and 61 received RIC with fludarabine and busulfan (FluBu) and GVHD prophylaxis with a calcineurin inhibitor and mycophenolate mofetil. The 2-year overall survival was superior in patients who received FCR versus FluBu (72.7% versus 54.1%, P = .031), and in multivariable analysis adjusted for Disease Risk Index and donor type, only the conditioning regimen (FluBu versus FCR: HR, 2.06; 95% CI, 1.04 to 4.08; P = .037) and Disease Risk Index (low versus intermediate/high: HR, .38; 95% CI, .17 to .86; P = .02) were independent predictors of overall survival. The 2-year cumulative incidence of chronic GVHD was lower in patients who received FCR versus FluBu (24.2% versus 51.7%, P = .01). There was no difference in rate of relapse/progression or acute GVHD. Our results demonstrate that the use of RIC with FCR and GVHD prophylaxis with a calcineurin inhibitor and mini-methotrexate is associated with decreased chronic GVHD and improved overall survival.
Collapse
Affiliation(s)
- Vanessa E Kennedy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Bipin N Savani
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplant, Department of Medicine, Veterans Affairs Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - John P Greer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Adetola A Kassim
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Brian G Engelhardt
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Stacey A Goodman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplant, Department of Medicine, Veterans Affairs Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Salyka Sengsayadeth
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Wichai Chinratanalab
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplant, Department of Medicine, Veterans Affairs Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Madan Jagasia
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Hematology/Oncology, Stem Cell Transplantation, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
94
|
Kim HT, Zhang MJ, Woolfrey AE, St Martin A, Chen J, Saber W, Perales MA, Armand P, Eapen M. Donor and recipient sex in allogeneic stem cell transplantation: what really matters. Haematologica 2016; 101:1260-1266. [PMID: 27354023 DOI: 10.3324/haematol.2016.147645] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 06/23/2016] [Indexed: 02/02/2023] Open
Abstract
We investigated whether and how recipient-donor sex affects transplantation outcomes of 11,797 patients transplanted between 2008 and 2010. Thirty-seven percent were male recipients with male donors, 21% male recipients with female donors, 25% female recipients with male donors, and 17% female recipients with female donors. In multivariable analyses, male recipients had inferior overall survival and progression-free survival compared to females regardless of donor sex, with an 11% relative increase in the hazard of death (P<0.0001) and a 10% relative increase in the hazard of death or relapse (P<0.0001). The detrimental effect of male recipients varied by donor sex. For male recipients with male donors, there was a 12% relative increase in the subdistribution hazard of relapse compared with female recipients with male donors (P=0.0036) and male recipients with female donors (P=0.0037). For male recipients with female donors, there was a 19% relative increase in the subdistribution hazard of non-relapse mortality compared with male recipients with male donors (P<0.0001) and a 22% relative increase compared with female recipients with male donors (P=0.0003). In addition, male recipients with female donors showed a 21% relative increase in the subdistribution hazard of chronic graft-versus-host disease (P<0.0001) compared with female recipients with male donors. Donor sex had no effect on outcomes for female recipients. Transplantation of grafts from male and female donors was associated with inferior overall survival and progression-free survival in male recipients with differing patterns of failure. Recipient sex is an important prognostic factor independent of donor sex.
Collapse
Affiliation(s)
- Haesook T Kim
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Mei-Jie Zhang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI
| | - Ann E Woolfrey
- Department of Medicine, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Andrew St Martin
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Junfang Chen
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Wael Saber
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | | | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mary Eapen
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
95
|
Grimstad F, Krieg S. Immunogenetic contributions to recurrent pregnancy loss. J Assist Reprod Genet 2016; 33:833-47. [PMID: 27169601 DOI: 10.1007/s10815-016-0720-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022] Open
Abstract
While sporadic pregnancy loss is common, occurring in 15 % of pregnancies, recurrent pregnancy loss (RPL) impacts approximately 5 % of couples. Though multiple causes are known (including structural, hormonal, infectious, autoimmune, and thrombophilic causes), after evaluation, roughly half of all cases remain unexplained. The idiopathic RPL cases pose a challenging therapeutic dilemma in addition to incurring much physical and emotional morbidity. Immunogenetic causes have been postulated to contribute to these cases of RPL. Natural Killer cell, T cell expression pattern changes in the endometrium have both been shown in patients with RPL. Human leukocyte antigen (HLA) and cytokine allelic variations have also been studied as etiologies for RPL. Some of the results have been promising, however the studies are small and have not yet put forth outcomes that would change our current diagnosis and management of RPL. Larger database studies are needed with stricter control criteria before reasonable conclusions can be drawn.
Collapse
Affiliation(s)
- Frances Grimstad
- Department of Obstetrics and Gynecology, University of Kansas, 3901 Rainbow Blvd MS 2028, Kansas City, KS, 66160, USA.
| | - Sacha Krieg
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Oregon Health Sciences University, Portland, OR, USA
| |
Collapse
|
96
|
Knorr DA, Wang H, Aurora M, MacMillan ML, Holtan SG, Bergerson R, Cao Q, Weisdorf DJ, Cooley S, Brunstein C, Miller JS, Wagner JE, Blazar BR, Verneris MR. Loss of T Follicular Helper Cells in the Peripheral Blood of Patients with Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2016; 22:825-33. [PMID: 26806586 PMCID: PMC5015683 DOI: 10.1016/j.bbmt.2016.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
Abstract
B cell antihost antibody production plays a central role in chronic graft-versus-host disease (cGVHD). T follicular helper (TFH) cells drive B cell responses and are implicated in this process. Given differences in cGVHD incidence between umbilical cord blood (UCB) and adult donor transplant recipients, we evaluated TFH cell reconstitution kinetics to define graft source differences and their potential pathogenic role in cGVHD. Although we observed significantly fewer TFH cells in the blood of UCB recipients (versus matched related donors [MRD]) early after transplantation, by 1 year the numbers of TFH cells were similar. Additionally, at both early (day 60) and late (1 year) time points, TFH cell phenotype was predominantly central memory cells in both cohorts. TFH cells were functional and able to produce multiple cytokines (INF-γ, TNF-α, IL-2, IL-17, and IL-21) after stimulation. In contrast to mouse models, where an enhanced frequency of splenic TFH cells contributes to cGVHD, patients with cGVHD showed significantly depleted circulating TFH cells after both UCB and MRD transplantation. Low numbers of TFH cells early after UCB transplantation could directly contribute to less cGVHD in this cohort. Additionally, systemic therapy (including steroids and calcineurin inhibitors) may contribute to decreases in TFH cells in patients with cGVHD. These data provide further evidence supporting the importance of TFH cells in cGVHD pathogenesis.
Collapse
Affiliation(s)
- David A Knorr
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Hongbo Wang
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Mukta Aurora
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Margaret L MacMillan
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Shernan G Holtan
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Rachel Bergerson
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Qing Cao
- Division of Blood and Marrow Transplantation, Department of Biostatistics, University of Minnesota, Minnesota
| | - Daniel J Weisdorf
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Sarah Cooley
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Claudio Brunstein
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - Jeffery S Miller
- Division of Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minnesota
| | - John E Wagner
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota
| | - Michael R Verneris
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minnesota.
| |
Collapse
|
97
|
Chronic graft-versus-host disease presenting as eosinophilic fasciitis: therapeutic challenges and an additional case. J Clin Rheumatol 2016; 21:86-94. [PMID: 25710860 DOI: 10.1097/rhu.0000000000000212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is one of the main late complications of allogeneic hematopoietic stem cell transplant and a major contributor to the mortality and morbidity in surviving recipients. Skin is the most common involved organ in cGVHD and may mimic a wide spectrum of dermatological conditions in its clinical and histopathologic manifestations. Some of the commonly simulated diseases are scleroderma, morphea, and lichen sclerosus. Chronic GVHD simulating eosinophilic fasciitis (EF) is relatively rare, frequently presenting with skin induration, a typical "peau d'orange" appearance, peripheral blood eosinophilia, myalgia, arthralgia, and arthritis leading to joint contractures in severe cases.Diagnosis is based on clinical manifestations and histopathology. Treatment is challenging because most cases are refractory to first-line therapy of glucocorticoids and calcineurin inhibitors (CNIs), and there is no standard second-line therapy.We report a comprehensive review of literature on all reported cases of CGVHD presenting as EF. We also describe an additional interesting case of cGVHD presenting as EF that was resistant to traditional therapy of high-dose glucocorticoids and cyclosporin A, but showed complete resolution of skin manifestations after addition of imatinib.Chronic GVHD presenting as EF is a rare variant of sclerodermatous cGVHD. Diagnosis is difficult, and treatment of cGVHD mimicking EF remains a therapeutic challenge because of obscure pathogenesis and poor response to traditional immunosuppressive medications. Emerging insights into the pathogenesis of cGVHD have resulted in the development of novel targeted therapies, which may improve outcomes and should be attempted in this subset of the disease. Larger studies are warranted to substantiate these preliminary findings.
Collapse
|
98
|
Circulating T follicular helper cells with increased function during chronic graft-versus-host disease. Blood 2016; 127:2489-97. [PMID: 26944544 DOI: 10.1182/blood-2015-12-688895] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) remains a major late complication of allogeneic hematopoietic stem cell transplantation (HSCT). Previous studies have established that both donor B and T cells contribute to immune pathology in cGVHD but the mechanisms responsible for coordinated B- and T-cell responses directed against recipient antigens have not been understood. T follicular helper cells (TFH) play an important role in the regulation of B-cell immunity. We performed extensive phenotypic and functional analysis of circulating TFH (cTFH) and B cells in 66 patients after HSCT. Patients with active cGVHD had a significantly lower frequency of cTFH compared with patients without cGVHD. This was associated with higher CXCL13 plasma levels suggesting increased homing of TFH to secondary lymphoid organs. In patients with active cGVHD, cTFH phenotype was skewed toward a highly activated profile with predominance of T helper 2 (Th2)/Th17 subsets. Activated cTFH in patients with cGVHD demonstrated increased functional ability to promote B-cell immunoglobulin secretion and maturation. Moreover, the activation signature of cTFH was highly correlated with increased B-cell activation and plasmablast maturation in patients after transplant. These studies provide new insights into the immune pathogenesis of human cGVHD and identify TFH as a key coordinating element supporting B-cell involvement in this disease.
Collapse
|
99
|
Desmarets M, Bardiaux L, Benzenine E, Dussaucy A, Binda D, Tiberghien P, Quantin C, Monnet E. Effect of storage time and donor sex of transfused red blood cells on 1-year survival in patients undergoing cardiac surgery: an observational study. Transfusion 2016; 56:1213-22. [DOI: 10.1111/trf.13537] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 01/10/2016] [Accepted: 01/12/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Maxime Desmarets
- Centre d'Investigation Clinique, INSERM CIC 1431; University Hospital of Besançon; Besançon France
| | - Laurent Bardiaux
- Etablissement Français du Sang (EFS) Pyrénées Méditerranée; Toulouse, France
| | - Eric Benzenine
- Service de Biostatistiques et Informatique Médicale (DIM), University Hospital of Dijon; Dijon, France
| | - Alain Dussaucy
- Département d'Information Médicale; University Hospital of Besançon
| | - Delphine Binda
- Centre d'Investigation Clinique, INSERM CIC 1431; University Hospital of Besançon; Besançon France
| | - Pierre Tiberghien
- UMR 1098, INSERM, Université de Franche-Comté, Etablissement Français du Sang; Besançon, France
| | - Catherine Quantin
- Service de Biostatistiques et Informatique Médicale (DIM), University Hospital of Dijon; Dijon, France
- Centre d'Investigation Clinique, INSERM CIC 1432, University Hospital of Dijon
- UMR 1181, Biostatistiques, biomathématiques, pharmacoépidémiologie et maladies infectieuses (BP2PHI), INSERM Université de Bourgogne; Dijon France
| | - Elisabeth Monnet
- Centre d'Investigation Clinique, INSERM CIC 1431; University Hospital of Besançon; Besançon France
- EA 4266, Agents Pathogènes et Inflammation, Université de Franche-Comté; Besançon France
| |
Collapse
|
100
|
Paul J, Sahaf B, Perloff S, Schoenrock K, Wu F, Nakasone H, Coller J, Miklos D. High-throughput allogeneic antibody detection using protein microarrays. J Immunol Methods 2016; 432:57-64. [PMID: 26902899 DOI: 10.1016/j.jim.2016.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 01/26/2023]
Abstract
Enzyme-linked immunosorbent assays (ELISAs) have traditionally been used to detect alloantibodies in patient plasma samples post hematopoietic cell transplantation (HCT); however, protein microarrays have the potential to be multiplexed, more sensitive, and higher throughput than ELISAs. Here, we describe the development of a novel and sensitive microarray method for detection of allogeneic antibodies against minor histocompatibility antigens encoded on the Y chromosome, called HY antigens. Six microarray surfaces were tested for their ability to bind recombinant protein and peptide HY antigens. Significant allogeneic immune responses were determined in male patients with female donors by considering normal male donor responses as baseline. HY microarray results were also compared with our previous ELISA results. Our overall goal was to maximize antibody detection for both recombinant protein and peptide epitopes. For detection of HY antigens, the Epoxy (Schott) protein microarray surface was both most sensitive and reliable and has become the standard surface in our microarray platform.
Collapse
Affiliation(s)
- Jed Paul
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA 94305, United States.
| | - Bita Sahaf
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA 94305, United States.
| | - Spenser Perloff
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA 94305, United States.
| | - Kelsi Schoenrock
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA 94305, United States
| | - Fang Wu
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA 94305, United States.
| | - Hideki Nakasone
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA 94305, United States.
| | - John Coller
- Stanford Functional Genomics Facility, Stanford University School of Medicine, 269 West Campus Dr., CCSR #0120, Stanford, CA 94305, United States.
| | - David Miklos
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA 94305, United States.
| |
Collapse
|