101
|
GLCCI1 and Glucocorticoid Receptor Genetic Diversity and Response to Glucocorticoid-Based Treatment of Graft-versus-Host Disease. Biol Blood Marrow Transplant 2015; 21:1246-50. [PMID: 25843653 DOI: 10.1016/j.bbmt.2015.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/16/2015] [Indexed: 11/20/2022]
Abstract
The genetic diversity of loci implicated in glucocorticoid (GC) response has been associated with interindividual variations in responsiveness to GC in various diseases, such as asthma and inflammatory bowel disorders. In acute graft-versus-host disease (aGVHD), similar differences of first-line therapy responsiveness are also observed, with approximately 40% of patients failing to respond to GC. Here, the distribution of functionally relevant single nucleotide polymorphisms (SNP) belonging to the GC-induced transcript 1 GLCCI1 (rs37972) and the glucocorticoid receptor (rs41423247, rs6195 and rs6198) gene loci were analyzed alongside clinical factors for their association with the response to corticosteroids in aGVHD. The frequencies of variant alleles did not differ significantly between corticoresistant patients, their donors, and their corticosensitive peers (P = .10 to 1.00). Severe and early onset of aGVHD, bone marrow as the stem cell source, and an HLA mismatch were associated with the failure to respond to GC in logistic regression. After including the single SNPs to the model, carriers of the rs41423247 polymorphism had a higher probability of responding to GC, whereas all other polymorphisms did not affect the likelihood of response.
Collapse
|
102
|
Lengliné E, Chevret S, Moreau AS, Pène F, Blot F, Bourhis JH, Buzyn A, Schlemmer B, Socié G, Azoulay E. Changes in intensive care for allogeneic hematopoietic stem cell transplant recipients. Bone Marrow Transplant 2015; 50:840-5. [PMID: 25798675 DOI: 10.1038/bmt.2015.55] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 12/14/2014] [Accepted: 12/28/2014] [Indexed: 01/01/2023]
Abstract
Intensive care unit (ICU) admission is associated with high mortality in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Whether mortality has decreased recently is unknown. The 497 adult allogeneic HSCT recipients admitted to three ICUs between 1997 and 2011 were evaluated retrospectively. Two hundred and nine patients admitted between 1997 and 2003 were compared with the 288 patients admitted from 2004 to 2011. Factors associated with 90-day mortality were identified. The recent cohort was characterized by older age, lower conditioning intensity, and greater use of peripheral blood or unrelated-donor graft. In the recent cohort, ICU was used more often for patients in hematological remission (67% vs 44%; P<0.0001) and without GVHD (73% vs 48%; P<0.0001) or invasive fungal infection (85% vs 73%; P=0.0003) despite a stable admission rate (21.7%). These changes were associated with significantly better 90-day survival (49% vs 31%). Independent predictors of hospital mortality were GVHD, mechanical ventilation (MV) and renal replacement therapy (RRT). Among patients who required MV or RRT, survival was 29% and 18%, respectively, but dropped to 18% and 6% in those with GVHD. The use of ICU admission has changed and translated into improved survival, but advanced life support in patients with GVHD usually provides no benefits.
Collapse
Affiliation(s)
- E Lengliné
- 1] Medical Intensive Care Unit, Hôpital Saint-Louis, AP-HP, Université Paris Diderot, Paris, France [2] Hematology, Immunology, Oncology Division (HOR), Hôpital Saint-Louis, AP-HP, Université Paris Diderot, Paris, France
| | - S Chevret
- 1] Biostatistic Unit, Hôpital Saint-Louis, AP-HP, Université Paris Diderot, Paris, France [2] ECSTRA team, CRESS (UMR 1153), Inserm, Université Paris Diderot Paris, France
| | - A-S Moreau
- Medical Intensive Care Unit, Hôpital Saint-Louis, AP-HP, Université Paris Diderot, Paris, France
| | - F Pène
- Medical Intensive Care Unit, Hôpital Cochin, AP-HP, Université Paris Descartes, Paris France
| | - F Blot
- Intensive Care Unit, Institut Gustave Rousy, Villejuif, France
| | - J-H Bourhis
- Hematology Department, Institut Gustave Roussy, Villejuif, France
| | - A Buzyn
- 1] Hematology Department, Hôpital Necker, AP-HP, Université Paris Descartes, Paris France [2] Institut National du Cancer, Boulogne Billancourt, France
| | - B Schlemmer
- Medical Intensive Care Unit, Hôpital Saint-Louis, AP-HP, Université Paris Diderot, Paris, France
| | - G Socié
- 1] Hematology, Immunology, Oncology Division (HOR), Hôpital Saint-Louis, AP-HP, Université Paris Diderot, Paris, France [2] INSERM UMR 1160, Paris France
| | - E Azoulay
- 1] Medical Intensive Care Unit, Hôpital Saint-Louis, AP-HP, Université Paris Diderot, Paris, France [2] ECSTRA team, CRESS (UMR 1153), Inserm, Université Paris Diderot Paris, France
| |
Collapse
|
103
|
Wang Z, Wang J. [Advances in prediction and diagnostic biomarkers on acute graft-versus-host disease after allogeneic stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:172-6. [PMID: 25778900 PMCID: PMC7342147 DOI: 10.3760/cma.j.issn.0253-2727.2015.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ziwei Wang
- Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jianmin Wang
- Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
104
|
High Number of Memory T Cells Is Associated with Higher Risk of Acute Graft-versus-Host Disease after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2015; 21:569-74. [DOI: 10.1016/j.bbmt.2014.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/11/2014] [Indexed: 11/18/2022]
|
105
|
Risk Factors, Pattern and Clinical Outcome of Acute Graft Versus Host Disease in Acute Leukemia Patients Undergoing Allogeneic Stem Cell Transplant. Indian J Hematol Blood Transfus 2015; 31:404-12. [PMID: 26306063 DOI: 10.1007/s12288-014-0499-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022] Open
Abstract
We sought to determine risk factors, pattern and outcome of acute graft versus host disease (aGVHD) in seventy-seven acute leukemia patients who underwent allogeneic stem cell transplant at our centre from January 2008 to March 2013. GVHD prophylaxis with cyclosporine-methotrexate or cyclosporine-mycophenolate mofetil was used. Patients were divided in 2 groups, grade II-IV aGVHD (group A) and grade 0-I aGVHD (group B). Incidence of any grade and grade II-IV aGVHD was 44 and 18 %, respectively. The most common site of aGVHD was gastro-intestinal tract (65 %) followed by skin (35 %). Higher total nucleated cell (TNC) dose infused was associated with increased incidence of grade II-IV aGVHD. Incidence of relapse and incidence of slippage of chimerism was 21 and 36 % in group A while 37 and 27 % in group B respectively. Transplant related mortality (TRM) was 21 % in group A and 13 % in group B. Probability of OS and RFS at 4 years was 63 and 34 % in group A compared with 40 and 38 % in group B, respectively. We conclude that higher TNC dose infused is a risk factor for grade II-IV aGVHD with gut being the commonest site. Grade II-IV aGVHD did not have a significant impact on incidence of relapse, TRM and OS.
Collapse
|
106
|
Holbro A, Stern M, Infanti L, O'Meara A, Drexler B, Frey BM, Tiercy JM, Passweg JR, Gassner C, Buser A, Sigle JP. Impact of recipient ABH secretor status on outcome in minor ABO-incompatible hematopoietic stem cell transplantation. Transfusion 2015; 55:64-9. [PMID: 24990435 DOI: 10.1111/trf.12768] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/19/2014] [Accepted: 05/22/2014] [Indexed: 11/27/2022]
Abstract
BACKGROUND The impact of ABO incompatibility on hematopoietic stem cell transplantation (HSCT) outcome is controversial. As ABH substances are expressed on tissues and secreted in body fluids, they could drive an immune response in minor ABO-incompatible HSCT. The aim of the study was to investigate the prognostic role of the recipients' ABH secretor status. STUDY DESIGN AND METHODS Patients who underwent minor ABO-incompatible HSCT were included. Secretor status was determined either serologically or by molecular genetics. RESULTS Between March 1996 and June 2012, a total of 176 patients received minor ABO-incompatible HSCT and 150 (85%) were secretors. Incidence and severity of acute graft-versus-host disease (GVHD) and chronic GVHD did not differ between secretors and nonsecretors (cumulative incidences ± standard errors: acute GVHD on Day 100, 41 ± 11 and 46 ± 5%, p = 0.59; chronic GVHD at 2 years, 52 ± 13 and 56 ± 5%, p = 0.62, for secretors and nonsecretors, respectively). Additionally, nonrelapse mortality (NRM) and overall survival (OS) were similar in the two groups (2-year NRM, 27 ± 9 and 23 ± 3%, p = 0.45; 4-year OS, 64 ± 10 and 55 ± 4%, p = 0.28, for secretors and nonsecretors, respectively). CONCLUSION The recipients' ABH secretor status in minor ABO-incompatible HSCT has no prognostic impact on major transplant outcomes.
Collapse
Affiliation(s)
- Andreas Holbro
- Division of Hematology, University Hospital, Basel, Switzerland; Blood Transfusion Centre, Swiss Red Cross, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Acute graft-versus-host disease of the kidney in allogeneic rat bone marrow transplantation. PLoS One 2014; 9:e115399. [PMID: 25541735 PMCID: PMC4277361 DOI: 10.1371/journal.pone.0115399] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 11/21/2014] [Indexed: 01/06/2023] Open
Abstract
Allogeneic hematopoietic cell or bone marrow transplantation (BMT) causes graft-versus-host-disease (GVHD). However, the involvement of the kidney in acute GVHD is not well-understood. Acute GVHD was induced in Lewis rats (RT1l) by transplantation of Dark Agouti (DA) rat (RT1(a)) bone marrow cells (6.0 × 10(7) cells) without immunosuppression after lethal irradiation (10 Gy). We examined the impact of acute GVHD on the kidney in allogeneic BMT rats and compared them with those in Lewis-to-Lewis syngeneic BMT control and non-BMT control rats. In syngeneic BMT and non-BMT control rats, acute GVHD did not develop by day 28. In allogeneic BMT rats, severe acute GVHD developed at 21-28 days after BMT in the skin, intestine, and liver with decreased body weight (>20%), skin rush, diarrhea, and liver dysfunction. In the kidney, infiltration of donor-type leukocytes was by day 28. Mild inflammation characterized by infiltration of CD3(+) T-cells, including CD8(+) T-cells and CD4(+) T-cells, and CD68(+) macrophages to the interstitium around the small arteries was noted. During moderate to severe inflammation, these infiltrating cells expanded into the peritubular interstitium with peritubular capillaritis, tubulitis, acute glomerulitis, and endarteritis. Renal dysfunction also developed, and the serum blood urea nitrogen (33.9 ± 4.7 mg/dL) and urinary N-acetyl-β-D-glucosaminidase (NAG: 31.5 ± 15.5 U/L) levels increased. No immunoglobulin and complement deposition was detected in the kidney. In conclusion, the kidney was a primary target organ of acute GVHD after BMT. Acute GVHD of the kidney was characterized by increased levels of urinary NAG and cell-mediated injury to the renal microvasculature and renal tubules.
Collapse
|
108
|
Li Y, Chen HL, Bannick N, Henry M, Holm AN, Metwali A, Urban JF, Rothman PB, Weiner GJ, Blazar BR, Elliott DE, Ince MN. Intestinal helminths regulate lethal acute graft-versus-host disease and preserve the graft-versus-tumor effect in mice. THE JOURNAL OF IMMUNOLOGY 2014; 194:1011-20. [PMID: 25527786 DOI: 10.4049/jimmunol.1303099] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Donor T lymphocyte transfer with hematopoietic stem cells suppresses residual tumor growth (graft-versus-tumor [GVT]) in cancer patients undergoing bone marrow transplantation (BMT). However, donor T cell reactivity to host organs causes severe and potentially lethal inflammation called graft-versus-host disease (GVHD). High-dose steroids or other immunosuppressive drugs are used to treat GVHD that have limited ability to control the inflammation while incurring long-term toxicity. Novel strategies are needed to modulate GVHD, preserve GVT, and improve the outcome of BMT. Regulatory T cells (Tregs) control alloantigen-sensitized inflammation of GVHD, sustain GVT, and prevent mortality in BMT. Helminths colonizing the alimentary tract dramatically increase the Treg activity, thereby modulating intestinal or systemic inflammatory responses. These observations led us to hypothesize that helminths can regulate GVHD and maintain GVT in mice. Acute GVHD was induced in helminth (Heligmosomoides polygyrus)-infected or uninfected BALB/c recipients of C57BL/6 donor grafts. Helminth infection suppressed donor T cell inflammatory cytokine generation and reduced GVHD-related mortality, but maintained GVT. H. polygyrus colonization promoted the survival of TGF-β-generating recipient Tregs after a conditioning regimen with total body irradiation and led to a TGF-β-dependent in vivo expansion/maturation of donor Tregs after BMT. Helminths did not control GVHD when T cells unresponsive to TGF-β-mediated immune regulation were used as donor T lymphocytes. These results suggest that helminths suppress acute GVHD using Tregs and TGF-β-dependent pathways in mice. Helminthic regulation of GVHD and GVT through intestinal immune conditioning may improve the outcome of BMT.
Collapse
Affiliation(s)
- Yue Li
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Hung-Lin Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Nadine Bannick
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Michael Henry
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Adrian N Holm
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Ahmed Metwali
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Joseph F Urban
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705; and
| | - Paul B Rothman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - George J Weiner
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | - David E Elliott
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - M Nedim Ince
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242;
| |
Collapse
|
109
|
Soriano R, Herrera S, Nogués X, Diez-Perez A. Current and future treatments of secondary osteoporosis. Best Pract Res Clin Endocrinol Metab 2014; 28:885-94. [PMID: 25432359 DOI: 10.1016/j.beem.2014.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Osteoporosis is commonly associated with menopause and ageing. It can, however, also be caused by diseases, lifestyle, genetic diseases, drug therapies and other therapeutic interventions. In cases of secondary osteoporosis, a common rule is the management of the underlying condition. Healthy habits and calcium and vitamin D supplementation are also generally advised. In cases of high risk of fracture, specific antiosteoporosis medications should be prescribed. For most conditions, the available evidence is limited. Special attention should be paid to possible contraindications of drugs used for the treatment of postmenopausal or senile osteoporosis. Bisphosphonates are the most widely used drugs in secondary osteoporosis, and denosumab or teriparatide have been also assessed in some cases. Important research is needed to develop more tailored strategies, specific to the peculiarities of the different types of secondary osteoporosis.
Collapse
Affiliation(s)
- Raquel Soriano
- Department of Internal Medicine, Hospital del Mar, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Spain; Autonomous University of Barcelona, Spain; RETICEF, Instituto Carlos III, Spain.
| | - Sabina Herrera
- Department of Internal Medicine, Hospital del Mar, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Spain; Autonomous University of Barcelona, Spain; RETICEF, Instituto Carlos III, Spain.
| | - Xavier Nogués
- Department of Internal Medicine, Hospital del Mar, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Spain; Autonomous University of Barcelona, Spain; RETICEF, Instituto Carlos III, Spain.
| | - Adolfo Diez-Perez
- Department of Internal Medicine, Hospital del Mar, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Spain; Autonomous University of Barcelona, Spain; RETICEF, Instituto Carlos III, Spain.
| |
Collapse
|
110
|
Brissot E, Bossard C, Malard F, Braudeau C, Chevallier P, Guillaume T, Delaunay J, Josien R, Gregoire M, Gaugler B, Mohty M. Involvement of the CX3CL1 (fractalkine)/CX3CR1 pathway in the pathogenesis of acute graft-versus-host disease. J Leukoc Biol 2014; 97:227-35. [PMID: 25420917 DOI: 10.1189/jlb.5hi0714-325r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study investigated the role of cytokines and chemokines in aGVHD incidence and severity in 109 patients who underwent reduced-intensity conditioning allogeneic stem cell transplantation (HSCT). Among the 42 cytokines tested at d 0 HSCT, only CX3CL1 levels at d 0 HSCT were significantly associated with Grades II-IV aGVHD development (P = 0.04). Increased levels of CX3CL1 at d 20-30 and 50 post-HSCT were also significantly associated with aGVHD (P = 0.02 and P = 0.03, respectively). No such association was found before the conditioning regimen or at d 100-120 post-HSCT. As the receptor for CX3CL1 is CX3CR1, the number of CX3CR1(+) cells was determined by flow cytometry. The CX3CR1(+)CD8(+) T cell proportion was significantly higher in patients with aGVHD than those without aGVHD (P = 0.01). To investigate the distribution of the CX3CL1/CX3CR1 axis in the anatomic sites of aGVHD, CX3CL1 and CX3CR1 levels were studied by use of an in situ immunohistochemical analysis on GI biopsies of patients with intestinal aGVHD. CX3CL1 expression was increased significantly in the epithelial cells and mononuclear cells of the lamina propria. CX3CR1(+) mononuclear cells were identified in close contact with epithelial cells. These findings strongly suggest the implication of the CX3CL1/CX3CR1 axis in the pathogenesis of aGVHD.
Collapse
Affiliation(s)
- Eolia Brissot
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Celine Bossard
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Florent Malard
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Cécile Braudeau
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Patrice Chevallier
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Thierry Guillaume
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jacques Delaunay
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Régis Josien
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marc Gregoire
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Beatrice Gaugler
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Mohamad Mohty
- *Universite Pierre et Marie Curie, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 938, Paris, France; Hématologie Clinique, Centre Hospitalier et Universitaire Hotel Dieu, Nantes, France; Le Centre Régional de Recherche en Cancérologie Nantes/Angers, Unité Mixte de Recherche 892 Institut National de la Santé et de la Recherche Médicale-6299 Centre National de la Recherche Scientifique, Nantes, France; EA4273 Biometadys, Faculté de Médecine, Université de Nantes, Nantes, France; Service d'Anatomie et Cytologie Pathologique and Laboratoire d'Immunologie, Plateforme d'Immuno-Monitorage Clinique, Centre Hospitalier et Universitaire de Nantes, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1098, Besançon, France; **Université de Franche-Comté, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France; Centre d'Investigation Clinique en Biothérapie CBT506, Plateforme de Biomonitoring, Besançon, France; and Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
111
|
Beloki L, Ciaurriz M, Mansilla C, Zabalza A, Perez-Valderrama E, Samuel ER, Lowdell MW, Ramirez N, Olavarria E. CMV-specific T cell isolation from G-CSF mobilized peripheral blood: depletion of myeloid progenitors eliminates non-specific binding of MHC-multimers. J Transl Med 2014; 12:317. [PMID: 25406933 PMCID: PMC4243324 DOI: 10.1186/s12967-014-0317-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/04/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cytomegalovirus (CMV)-specific T cell infusion to immunocompromised patients following allogeneic Hematopoietic Stem Cell Transplantation (allo-HSCT) is able to induce a successful anti-viral response. These cells have classically been manufactured from steady-state apheresis samples collected from the donor in an additional harvest prior to G-CSF mobilization, treatment that induces hematopoietic stem cell (HSC) mobilization to the periphery. However, two closely-timed cellular collections are not usually available in the unrelated donor setting, which limits the accessibility of anti-viral cells for adoptive immunotherapy. CMV-specific cytotoxic T cell (CTL) manufacture from the same G-CSF mobilized donor stem cell harvest offers great regulatory advantages, but the isolation using MHC-multimers is hampered by the high non-specific binding to myeloid progenitors, which reduces the purity of the cellular product. METHODS In the present study we describe an easy and fast method based on plastic adherence to remove myeloid cell subsets from 11 G-CSF mobilized donor samples. CMV-specific CTLs were isolated from the non-adherent fraction using pentamers and purity and yield of the process were compared to products obtained from unmanipulated samples. RESULTS After the elimination of unwanted cell subtypes, non-specific binding of pentamers was notably reduced. Accordingly, following the isolation process the purity of the obtained cellular product was significantly improved. CONCLUSIONS G-CSF mobilized leukapheresis samples can successfully be used to isolate antigen-specific T cells with MHC-multimers to be adoptively transferred following allo-HSCT, widening the accessibility of this therapy in the unrelated donor setting. The combination of the clinically translatable plastic adherence process to the antigen-specific cell isolation using MHC-multimers improves the quality of the therapeutic cellular product, thereby reducing the clinical negative effects associated with undesired alloreactive cell infusion.
Collapse
Affiliation(s)
- Lorea Beloki
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, Irunlarrea 3, 31008, Pamplona, Spain.
| | - Miriam Ciaurriz
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, Irunlarrea 3, 31008, Pamplona, Spain.
| | - Cristina Mansilla
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, Irunlarrea 3, 31008, Pamplona, Spain.
| | - Amaya Zabalza
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, Irunlarrea 3, 31008, Pamplona, Spain.
| | - Estela Perez-Valderrama
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, Irunlarrea 3, 31008, Pamplona, Spain.
| | - Edward R Samuel
- Department of Haematology, University College London Medical School, University College London, London, UK.
| | - Mark W Lowdell
- Department of Haematology, University College London Medical School, University College London, London, UK.
| | - Natalia Ramirez
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, Irunlarrea 3, 31008, Pamplona, Spain.
| | - Eduardo Olavarria
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, Irunlarrea 3, 31008, Pamplona, Spain. .,Department of Haematology, Complejo Hospitalario de Navarra, Navarra Health Service, Pamplona, Spain.
| |
Collapse
|
112
|
Abstract
n this issue of Blood, Hazenberg and Spits provide a detailed overview of human innate lymphoid cell (ILC) subsets and their development and distribution throughout the human body, discussing these cells in the context of human disease. In the same issue, Munneke et al for the first time link ILCs to human hematopoietic malignancies by identifying a clear correlation between the presence of activated ILCs after induction chemotherapy and the absence of acute graft-versus-host disease (GVHD) development following subsequent hematopoietic stem cell transplantation (HSCT).
Collapse
|
113
|
Hematopoietic stem cell transplantation: graft versus host disease and pathology of gastrointestinal tract, liver, and lung. Adv Anat Pathol 2014; 21:301-20. [PMID: 25105933 DOI: 10.1097/pap.0000000000000032] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hematopoietic stem cell transplantation (HCT), formerly known as bone marrow transplantation, is an integral part of treatment for many hematological malignancies. HCT is associated with several complications and comorbidities with differential effects on a wide spectrum of organs and tissues. We present an update on HCT-associated complications such as graft versus host disease (GVHD) and infection, with focus on the surgical pathology of the gastrointestinal (GI) tract, liver, and lung. Although the grading system for GI tract acute GVHD was proposed 40 years ago, recent studies have shed light on minimal histologic criteria for diagnosis of GVHD, as well as its differential diagnosis, including histologic effects of various medications. GI dysfunction in autologous transplant recipients is increasingly appreciated and patients are often biopsied. Acute liver injury in HCT is often due to sinusoidal obstruction syndrome (previously known as venoocclusive disease), or acute GVHD. Liver dysfunction at later time posttransplantation may be associated with acute or chronic GVHD, iron overload, or other causes of hepatitis. Lung injury in HCT is multifactorial, and it remains crucially important to diagnose and treat pulmonary infections. The pulmonary biopsy yields clinically unsuspected diagnoses in the majority of cases and its utilization is likely to increase. The pathology of the skin and kidney in HCT patients are detailed in accompanying articles.
Collapse
|
114
|
Donor CD4+ Foxp3+ regulatory T cells are necessary for posttransplantation cyclophosphamide-mediated protection against GVHD in mice. Blood 2014; 124:2131-41. [PMID: 25139358 DOI: 10.1182/blood-2013-10-525873] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Posttransplantation cyclophosphamide (PTCy) is an effective prophylaxis against graft-versus-host disease (GVHD). However, it is unknown whether PTCy works singularly by eliminating alloreactive T cells via DNA alkylation or also by restoring the conventional (Tcon)/regulatory (Treg) T-cell balance. We studied the role of Tregs in PTCy-mediated GVHD prophylaxis in murine models of allogeneic blood or marrow transplantation (alloBMT). In 2 distinct MHC-matched alloBMT models, infusing Treg-depleted allografts abrogated the GVHD-prophylactic activity of PTCy. Using allografts in which Foxp3(+) Tregs could be selectively depleted in vivo, either pre- or post-PTCy ablation of donor thymus-derived Tregs (tTregs) abolished PTCy protection against GVHD. PTCy treatment was associated with relative preservation of donor Tregs. Experiments using combinations of Foxp3(-) Tcons and Foxp3(+) Tregs sorted from different Foxp3 reporter mice indicated that donor Treg persistence after PTCy treatment was predominantly caused by survival of functional tTregs that retained Treg-specific demethylation and also induction of peripherally derived Tregs. Finally, adoptive transfer of tTregs retrieved from PTCy-treated chimeras rescued PTCy-treated, Treg-depleted recipients from lethal GVHD. Our findings indicate that PTCy-mediated protection against GVHD is not singularly dependent on depletion of donor alloreactive T cells but also requires rapidly recovering donor Tregs to initiate and maintain alloimmune regulation.
Collapse
|
115
|
De Bock M, Beguin Y, Leprince P, Willems E, Baron F, Deroyer C, Seidel L, Cavalier E, de Seny D, Malaise M, Gothot A, Merville MP, Fillet M. Comprehensive plasma profiling for the characterization of graft-versus-host disease biomarkers. Talanta 2014; 125:265-75. [DOI: 10.1016/j.talanta.2014.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 02/28/2014] [Accepted: 03/11/2014] [Indexed: 02/07/2023]
|
116
|
Lepore M, de Lalla C, Gundimeda SR, Gsellinger H, Consonni M, Garavaglia C, Sansano S, Piccolo F, Scelfo A, Häussinger D, Montagna D, Locatelli F, Bonini C, Bondanza A, Forcina A, Li Z, Ni G, Ciceri F, Jenö P, Xia C, Mori L, Dellabona P, Casorati G, De Libero G. A novel self-lipid antigen targets human T cells against CD1c(+) leukemias. ACTA ACUST UNITED AC 2014; 211:1363-77. [PMID: 24935257 PMCID: PMC4076585 DOI: 10.1084/jem.20140410] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
T cells that recognize self-lipids presented by CD1c are frequent in the peripheral blood of healthy individuals and kill transformed hematopoietic cells, but little is known about their antigen specificity and potential antileukemia effects. We report that CD1c self-reactive T cells recognize a novel class of self-lipids, identified as methyl-lysophosphatidic acids (mLPAs), which are accumulated in leukemia cells. Primary acute myeloid and B cell acute leukemia blasts express CD1 molecules. mLPA-specific T cells efficiently kill CD1c(+) acute leukemia cells, poorly recognize nontransformed CD1c-expressing cells, and protect immunodeficient mice against CD1c(+) human leukemia cells. The identification of immunogenic self-lipid antigens accumulated in leukemia cells and the observed leukemia control by lipid-specific T cells in vivo provide a new conceptual framework for leukemia immune surveillance and possible immunotherapy.
Collapse
Affiliation(s)
- Marco Lepore
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudia de Lalla
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - S Ramanjaneyulu Gundimeda
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Heiko Gsellinger
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Michela Consonni
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudio Garavaglia
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sebastiano Sansano
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Francesco Piccolo
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Scelfo
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniel Häussinger
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Daniela Montagna
- Laboratorio di Immunologia, Dipartimento di Pediatria, Università di Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology-Oncology, IRCCS Bambino Gesù Hospital, 00165 Rome, Italy
| | - Chiara Bonini
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Attilio Bondanza
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandra Forcina
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Zhiyuan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Guanghui Ni
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Fabio Ciceri
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Paul Jenö
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Chengfeng Xia
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore 138648
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore 138648
| |
Collapse
|
117
|
Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood 2014; 124:812-21. [PMID: 24855210 DOI: 10.1182/blood-2013-11-536888] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is widely used to treat hematopoietic cell disorders but is often complicated by graft-versus-host disease (GVHD), which causes severe epithelial damage. Here we have investigated longitudinally the effects of induction chemotherapy, conditioning radiochemotherapy, and allogeneic HSCT on composition, phenotype, and recovery of circulating innate lymphoid cells (ILCs) in 51 acute leukemia patients. We found that reconstitution of ILC1, ILC2, and NCR(-)ILC3 was slow compared with that of neutrophils and monocytes. NCR(+) ILC3 cells, which are not present in the circulation of healthy persons, appeared both after induction chemotherapy and after allogeneic HSCT. Circulating patient ILCs before transplantation, as well as donor ILCs after transplantation, expressed activation (CD69), proliferation (Ki-67), and tissue homing markers for gut (α4β7, CCR6) and skin (CCR10 and CLA). The proportion of ILCs expressing these markers was associated with a decreased susceptibility to therapy-induced mucositis and acute GVHD. Taken together, these data suggest that ILC recovery and treatment-related tissue damage are interrelated and affect the development of GVHD.
Collapse
|
118
|
Padet L, Loubaki L, Bazin R. Induction of PD-L1 on monocytes: a new mechanism by which IVIg inhibits mixed lymphocyte reactions. Immunobiology 2014; 219:687-94. [PMID: 24875729 DOI: 10.1016/j.imbio.2014.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/25/2014] [Accepted: 04/28/2014] [Indexed: 01/25/2023]
Abstract
Allograft rejection and graft-versus-host disease (GvHD) are frequent complications following solid organ or stem cell transplantation in which T cell activation plays a central role. Despite the development of new immunosuppressive drugs that improve the success rate of transplantation, allograft survival continues to be a challenge. Recently, intravenous immunoglobulin (IVIg) has been proposed as prophylaxis and post-transplant treatment to reduce acute rejection episodes. IVIg is a therapeutic agent that is known to down-modulate T cell functions in patients with autoimmune disorders. To test the hypothesis that this immunomodulatory effect could be beneficial in the context of transplantation, we used mixed lymphocyte reactions (MLR) as an in vitro model of allograft rejection and GvHD. Our results show that IVIg strongly inhibits the MLR as evaluated by IL-2 secretion, a well-known marker of T cell activation. IVIg also modulates the secretion of other pro-(IL-6, IFN-γ) and anti-inflammatory (IL-1RA) cytokines. More importantly, we show that IVIg induces monocytes with a CD80(low) PD-L1(high) phenotype and that blockade of PD-L1 partially abrogates the inhibitory effect of IVIg. We have thus identified a new mechanism by which IVIg inhibits T cell functions in the context of transplantation, supporting the potential usefulness of IVIg in the prevention or treatment of graft rejection and GvHD.
Collapse
Affiliation(s)
- Lauriane Padet
- Department of Research and Development, Héma-Québec, Québec, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec, Canada
| | - Lionel Loubaki
- Department of Research and Development, Héma-Québec, Québec, Canada
| | - Renée Bazin
- Department of Research and Development, Héma-Québec, Québec, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec, Canada.
| |
Collapse
|
119
|
Yeh AC, Brunner AM, Spitzer TR, Chen YB, Coughlin E, McAfee S, Ballen K, Attar E, Caron M, Preffer FI, Yeap BY, Dey BR. Phase I Study of Urate Oxidase in the Reduction of Acute Graft-Versus-Host Disease after Myeloablative Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2014; 20:730-4. [DOI: 10.1016/j.bbmt.2014.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 02/04/2014] [Indexed: 12/19/2022]
|
120
|
Ma Q, Li D, Carreño R, Patenia R, Tsai KY, Xydes-Smith M, Alousi AM, Champlin RE, Sale GE, Afshar-Kharghan V. Complement component C3 mediates Th1/Th17 polarization in human T-cell activation and cutaneous GVHD. Bone Marrow Transplant 2014; 49:972-6. [PMID: 24777193 DOI: 10.1038/bmt.2014.75] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 02/19/2014] [Accepted: 02/22/2014] [Indexed: 12/19/2022]
Abstract
The complement system has been shown to regulate T-cell activation and alloimmune responses in GVHD. Mice deficient in the central component of complement system C3 have significantly lower GVHD-related mortality/morbidity, and C3 modulates Th1/Th17 polarization in mouse GVHD. To investigate whether anticomplement therapy has any impact on human T-cell activation, a drug candidate Compstatin was used to inhibit C3 activation in this study. We found the frequency of IFN-γ (Th1)-, IL-4 (Th2)-, IL-17 (Th17)-, IL-2- and TNF-α-producing cells were significantly reduced among activated CD4(+) cells in the presence of Compstatin. Compstatin treatment decreased the proliferation of both CD4(+) and CD8(+) T cells upon TCR stimulation. However, Compstatin does not affect the production of IL-2 and TNF-α in activated CD8(+) T cells, and the differentiation of CD8(+) T cells into distinct memory and effector subsets remained intact. Furthermore, we examined complement deposition in skin and lip biopsy samples of patients diagnosed with cutaneous GVHD. C3 deposition was detected in the squamous epithelium and dermis, blood vessels and damaged sweat glands, and was associated with gland damage and regeneration. We conclude that C3 mediates Th1/Th17 polarization in human T-cell activation and skin GVHD in patients.
Collapse
Affiliation(s)
- Q Ma
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D Li
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R Carreño
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R Patenia
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K Y Tsai
- 1] Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, TX, USA [2] Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Xydes-Smith
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G E Sale
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - V Afshar-Kharghan
- Department of Benign Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
121
|
Ju JM, Lee H, Oh K, Lee DS, Choi EY. Kinetics of IFN-γ and IL-17 Production by CD4 and CD8 T Cells during Acute Graft-versus-Host Disease. Immune Netw 2014; 14:89-99. [PMID: 24851098 PMCID: PMC4022783 DOI: 10.4110/in.2014.14.2.89] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/16/2014] [Accepted: 03/27/2014] [Indexed: 02/07/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a fatal complication that occurs after allogeneic hematopoietic stem cell transplantation. To understand the dynamics of CD4 and CD8 T cell production of IFN-γ and IL-17 during GVHD progression, we established a GVHD model by transplanting T cell-depleted bone marrow (TCD-BM) and purified T cells from B6 mice into irradiated BALB.B, creating an MHC-matched but minor histocompatibility (H) antigen-mismatched transplantation (B6 → BALB.B GVHD). Transplantation-induced GVHD was confirmed by the presence of the appropriate compositional changes in the T cell compartments and innate immune cells in the blood and the systemic secretion of inflammatory cytokines. Using this B6 → BALB.B GVHD model, we showed that the production of IFN-γ and IL-17 by CD4 T cells preceded that by CD8 T cells in the spleen, mesenteric lymph node, liver, and lung in the BALB.B GVHD host, and Th1 differentiation predated Th17 differentiation in all organs during GVHD progression. Such changes in cytokine production were based on changes in cytokine gene expression by the T cells at different time points during GVHD development. These results demonstrate that both IFN-γ and IL-17 are produced by CD4 and CD8 T cells but with different kinetics during GVHD progression.
Collapse
Affiliation(s)
- Ji-Min Ju
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Hakmo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul 110-799, Korea
| | - Keunhee Oh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| |
Collapse
|
122
|
Fu J, Heinrichs J, Yu XZ. Helper T-cell differentiation in graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Arch Immunol Ther Exp (Warsz) 2014; 62:277-301. [PMID: 24699629 DOI: 10.1007/s00005-014-0284-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 01/27/2014] [Indexed: 02/07/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an effective therapeutic option for many malignant diseases. However, the efficacy of allo-HSCT is limited by the occurrence of destructive graft-versus-host disease (GVHD). Since allogeneic T cells are the driving force in the development of GVHD, their activation, proliferation, and differentiation are key factors to understanding GVHD pathogenesis. This review focuses on one critical aspect: the differentiation and function of helper T (Th) cells in acute GVHD. We first summarize well-established subsets including Th1, Th2, Th17, and T-regulatory cells; their flexibility, plasticity, and epigenetic modification; and newly identified subsets including Th9, Th22, and T follicular helper cells. Next, we extensively discuss preclinical findings of Th-cell lineages in GVHD: the networks of transcription factors involved in differentiation, the cytokine and signaling requirements for development, the reciprocal differentiation features, and the regulation of microRNAs on T-cell differentiation. Finally, we briefly summarize the recent findings on the roles of T-cell subsets in clinical GVHD and ongoing strategies to modify T-cell differentiation for controlling GVHD in patients. We believe further exploration and understanding of the immunobiology of T-cell differentiation in GVHD will expand therapeutic options for the continuing success of allo-HSCT.
Collapse
Affiliation(s)
- Jianing Fu
- Cancer Biology PhD Program, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL, 33612, USA
| | | | | |
Collapse
|
123
|
Minimal change disease in graft versus host disease: a podocyte response to the graft? Clin Nephrol 2014; 80:469-73. [PMID: 23006339 PMCID: PMC4504137 DOI: 10.5414/cn107420] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2013] [Indexed: 12/17/2022] Open
Abstract
Nephrotic syndrome is a rare complication of hematopoietic cell transplantation. It has been suggested that nephrotic syndrome may represent a limited form of graft-versus-host disease although the pathological link between these two entities remains unclear. In this paper, we report a case of a 61-year-old female who underwent nonmyeloablative allogenic stem cell transplantation for T-cell prolymphocytic leukemia and subsequently developed biopsy proven minimal change disease shortly after cessation of her immunosuppression therapy. Urinary CD80 was markedly elevated during active disease and disappeared following corticosteroid-induced remission. We hypothesize that alloreactive donor T cells target the kidney and induce podocyte expression of CD80 that results in proteinuria from limited 'graft versus host' disease.
Collapse
|
124
|
Singer BD, King LS, D'Alessio FR. Regulatory T cells as immunotherapy. Front Immunol 2014; 5:46. [PMID: 24575095 PMCID: PMC3920065 DOI: 10.3389/fimmu.2014.00046] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 01/27/2014] [Indexed: 12/23/2022] Open
Abstract
Regulatory T cells (Tregs) suppress exuberant immune system activation and promote immunologic tolerance. Because Tregs modulate both innate and adaptive immunity, the biomedical community has developed an intense interest in using Tregs for immunotherapy. Conditions that require clinical tolerance to improve outcomes – autoimmune disease, solid organ transplantation, and hematopoietic stem cell transplantation – may benefit from Treg immunotherapy. Investigators have designed ex vivo strategies to isolate, preserve, expand, and infuse Tregs. Protocols to manipulate Treg populations in vivo have also been considered. Barriers to clinically feasible Treg immunotherapy include Treg stability, off-cell effects, and demonstration of cell preparation purity and potency. Clinical trials involving Treg adoptive transfer to treat graft versus host disease preliminarily demonstrated the safety and efficacy of Treg immunotherapy in humans. Future work will need to confirm the safety of Treg immunotherapy and establish the efficacy of specific Treg subsets for the treatment of immune-mediated disease.
Collapse
Affiliation(s)
- Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University , Baltimore, MD , USA
| | - Landon S King
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University , Baltimore, MD , USA
| | - Franco R D'Alessio
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|
125
|
Krummey SM, Floyd TL, Liu D, Wagener ME, Song M, Ford ML. Candida-elicited murine Th17 cells express high Ctla-4 compared with Th1 cells and are resistant to costimulation blockade. THE JOURNAL OF IMMUNOLOGY 2014; 192:2495-504. [PMID: 24493820 DOI: 10.4049/jimmunol.1301332] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Effector and memory T cells may cross-react with allogeneic Ags to mediate graft rejection. Whereas the costimulation properties of Th1 cells are well studied, relatively little is known about the costimulation requirements of microbe-elicited Th17 cells. The costimulation blocker CTLA-4 Ig has been ineffective in the treatment of several Th17-driven autoimmune diseases and is associated with severe acute rejection following renal transplantation, leading us to investigate whether Th17 cells play a role in CD28/CTLA-4 blockade-resistant alloreactivity. We established an Ag-specific model in which Th1 and Th17 cells were elicited via Mycobacterium tuberculosis and Candida albicans immunization, respectively. C. albicans immunization elicited a higher frequency of Th17 cells and conferred resistance to costimulation blockade following transplantation. Compared with the M. tuberculosis group, C. albicans-elicited Th17 cells contained a higher frequency of IL-17(+)IFN-γ(+) producers and a lower frequency of IL-10(+) and IL-10(+)IL-17(+) cells. Importantly, Th17 cells differentially regulated the CD28/CTLA-4 pathway, expressing similarly high CD28 but significantly greater amounts of CTLA-4 compared with Th1 cells. Ex vivo blockade experiments demonstrated that Th17 cells are more sensitive to CTLA-4 coinhibition and therefore less susceptible to CTLA-4 Ig. These novel insights into the differential regulation of CTLA-4 coinhibition on CD4(+) T cells have implications for the immunomodulation of pathologic T cell responses during transplantation and autoimmunity.
Collapse
Affiliation(s)
- Scott M Krummey
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30322
| | | | | | | | | | | |
Collapse
|
126
|
Intravital imaging of donor allogeneic effector and regulatory T cells with host dendritic cells during GVHD. Blood 2014; 123:1604-14. [PMID: 24415540 DOI: 10.1182/blood-2013-09-526020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a systemic inflammatory response due to the recognition of major histocompatibility complex disparity between donor and recipient after hematopoietic stem cell transplantation (HSCT). T-cell activation is critical to the induction of GVHD, and data from our group and others have shown that regulatory T cells (Tregs) prevent GVHD when given at the time of HSCT. Using multiphoton laser scanning microscopy, we examined the single cell dynamics of donor T cells and dendritic cells (DCs) with or without Tregs postallogeneic transplantation. We found that donor conventional T cells (Tcons) spent very little time screening host DCs. Tcons formed stable contacts with DCs very early after transplantation and only increased velocity in the lymph node at 20 hours after transplant. We also observed that Tregs reduced the interaction time between Tcons and DCs, which was dependent on the generation of interleukin 10 by Tregs. Imaging using inducible Tregs showed similar disruption of Tcon-DC contact. Additionally, we found that donor Tregs induce host DC death and down-regulate surface proteins required for donor T-cell activation. These data indicate that Tregs use multiple mechanisms that affect host DC numbers and function to mitigate acute GVHD.
Collapse
|
127
|
|
128
|
Al-Homsi AS, Lai Z, Roy TS, Al-Malki MM, Kouttab N, Junghans RP. Post-transplant cyclophosphamide and bortezomib inhibit dendritic cell maturation and function and alter their IκB and NFκB. Transpl Immunol 2014; 30:40-5. [DOI: 10.1016/j.trim.2013.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/10/2013] [Accepted: 11/11/2013] [Indexed: 11/30/2022]
|
129
|
Bürgler D, Medinger M, Passweg J, Fischmann A, Bucher C. Intra-arterial catheter guided steroid administration for the treatment of steroid-refractory intestinal GvHD. Leuk Res 2013; 38:184-7. [PMID: 24268317 DOI: 10.1016/j.leukres.2013.10.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/01/2013] [Accepted: 10/26/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Acute gastrointestinal GvHD (GI-aGvHD) refractory to first-line treatment with systemic corticosteroids is resulting in death in the majority of patients. We prospectively assessed the feasibility and efficacy of regional intra-arterial steroid treatment in adult patients with severe (≥ grade III) GI-aGvHD not responding to first-line treatment. PATIENTS AND METHODS Patients with more than +++ GI-aGvHD not responding to intravenous methylprednisolone at a dose of 2mg/kg/day were eligible for inclusion. Catheter guided intra-arterial steroid administration (IASA) was performed into the superior and inferior mesenteric artery. RESULTS 12 consecutive patients with steroid-refractory grade III GI-aGvHD received IASA as second-line treatment. 83% of patients had gastrointestinal response including four patients (33%) with complete response at 28 days after IASA. 5/12 patients were alive at a median time of 531 days. CONCLUSION Regional treatment of severe GVHD with IASA treatment seems to be a safe and effective second-line treatment for steroid-refractory GI-aGvHD in adult patients.
Collapse
Affiliation(s)
- David Bürgler
- Clinic of Radiology and Nuclear Medicine, University Hospital Basel, Switzerland
| | - Michael Medinger
- Stem Cell Transplant Team, Division of Hematology, University Hospital Basel, Switzerland.
| | - Jakob Passweg
- Stem Cell Transplant Team, Division of Hematology, University Hospital Basel, Switzerland
| | - Arne Fischmann
- Clinic of Radiology and Nuclear Medicine, University Hospital Basel, Switzerland
| | - Christoph Bucher
- Stem Cell Transplant Team, Division of Hematology, University Hospital Basel, Switzerland
| |
Collapse
|
130
|
Saha A, Aoyama K, Taylor PA, Koehn BH, Veenstra RG, Panoskaltsis-Mortari A, Munn DH, Murphy WJ, Azuma M, Yagita H, Fife BT, Sayegh MH, Najafian N, Socie G, Ahmed R, Freeman GJ, Sharpe AH, Blazar BR. Host programmed death ligand 1 is dominant over programmed death ligand 2 expression in regulating graft-versus-host disease lethality. Blood 2013; 122:3062-73. [PMID: 24030385 PMCID: PMC3811178 DOI: 10.1182/blood-2013-05-500801] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 09/03/2013] [Indexed: 12/15/2022] Open
Abstract
Programmed death 1 (PD-1) and its ligands, PD-L1 and PD-L2, play an important role in the maintenance of peripheral tolerance. We explored the role of PD-1 ligands in regulating graft-versus-host disease (GVHD). Both PD-L1 and PD-L2 expression were upregulated in the spleen, liver, colon, and ileum of GVHD mice. Whereas PD-L2 expression was limited to hematopoietic cells, hematopoietic and endothelial cells expressed PD-L1. PD-1/PD-L1, but not PD-1/PD-L2, blockade markedly accelerated GVHD-induced lethality. Chimera studies suggest that PD-L1 expression on host parenchymal cells is more critical than hematopoietic cells in regulating acute GVHD. Rapid mortality onset in PD-L1-deficient hosts was associated with increased gut T-cell homing and loss of intestinal epithelial integrity, along with increased donor T-cell proliferation, activation, Th1 cytokine production, and reduced apoptosis. Bioenergetics profile analysis of proliferating alloreactive donor T-cells demonstrated increased aerobic glycolysis and oxidative phosphorylation in PD-L1-deficient hosts. Donor T-cells exhibited a hyperpolarized mitochondrial membrane potential, increased superoxide production, and increased expression of a glucose transporter in PD-L1-deficient hosts. Taken together, these data provide new insight into the differential roles of host PD-L1 and PD-L2 and their associated cellular and metabolic mechanisms controlling acute GVHD.
Collapse
Affiliation(s)
- Asim Saha
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Zhang P, Tey SK, Koyama M, Kuns RD, Olver SD, Lineburg KE, Lor M, Teal BE, Raffelt NC, Raju J, Leveque L, Markey KA, Varelias A, Clouston AD, Lane SW, MacDonald KPA, Hill GR. Induced regulatory T cells promote tolerance when stabilized by rapamycin and IL-2 in vivo. THE JOURNAL OF IMMUNOLOGY 2013; 191:5291-303. [PMID: 24123683 DOI: 10.4049/jimmunol.1301181] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Natural regulatory T cells (nTregs) play an important role in tolerance; however, the small numbers of cells obtainable potentially limit the feasibility of clinical adoptive transfer. Therefore, we studied the feasibility and efficacy of using murine-induced regulatory T cells (iTregs) for the induction of tolerance after bone marrow transplantation. iTregs could be induced in large numbers from conventional donor CD4 and CD8 T cells within 1 wk and were highly suppressive. During graft-versus-host disease (GVHD), CD4 and CD8 iTregs suppressed the proliferation of effector T cells and the production of proinflammatory cytokines. However, unlike nTregs, both iTreg populations lost Foxp3 expression within 3 wk in vivo, reverted to effector T cells, and exacerbated GVHD. The loss of Foxp3 in iTregs followed homeostatic and/or alloantigen-driven proliferation and was unrelated to GVHD. However, the concurrent administration of rapamycin, with or without IL-2/anti-IL-2 Ab complexes, to the transplant recipients significantly improved Foxp3 stability in CD4 iTregs (and, to a lesser extent, CD8 iTregs), such that they remained detectable 12 wk after transfer. Strikingly, CD4, but not CD8, iTregs could then suppress Teff proliferation and proinflammatory cytokine production and prevent GVHD in an equivalent fashion to nTregs. However, at high numbers and when used as GVHD prophylaxis, Tregs potently suppress graft-versus-leukemia effects and so may be most appropriate as a therapeutic modality to treat GVHD. These data demonstrate that CD4 iTregs can be produced rapidly in large, clinically relevant numbers and, when transferred in the presence of systemic rapamycin and IL-2, induce tolerance in transplant recipients.
Collapse
Affiliation(s)
- Ping Zhang
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Effect of novel proteasome and immunoproteasome inhibitors on dendritic cell maturation, function, and expression of IκB and NFκB. Transpl Immunol 2013; 29:1-6. [PMID: 24103732 DOI: 10.1016/j.trim.2013.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/24/2013] [Accepted: 09/27/2013] [Indexed: 11/23/2022]
Abstract
Dendritic cells (DC) play a central role in the pathophysiology of graft versus host disease (GvHD). Their antigen presenting capacity is nuclear factor κB- (NF-κB) dependent. Consequently, DC have emerged as a potential target for the prevention of GvHD and clinical trials with bortezomib are underway. We explored the activity of novel proteasome and immunoproteasome inhibitors on healthy volunteer peripheral blood DC. After incubation with the drug or drug combination, DC were stimulated with lipopolysaccharide, stained for maturation surface markers and then analyzed by flow cytometry. We found that the different molecule(s) inhibited DC maturation marker expression to variable degrees, with the constitutive proteasome-selective agent being the least active. In a DC and allogeneic CD4+ mixed lymphocyte reaction, DC incubation with the studied proteasome and immunoproteasome inhibitor(s), impeded T cell proliferation as measured by BrDU incorporation. Finally, we found that DC incubation with the drug(s) enhanced IκB expression and that oprozomib inhibited NF-κB expression. We concluded that based on its activity and oral bioavailability, oprozomib merits further investigation in an animal GvHD prevention model. We also suggest that altering IκB and NF-κB expressions may, in DC, represent a new mechanism of action of proteasome and immunoproteasome inhibitors.
Collapse
|
133
|
Inamoto Y, Martin PJ, Storer BE, Mielcarek M, Storb RF, Carpenter PA. Response endpoints and failure-free survival after initial treatment for acute graft-versus-host disease. Haematologica 2013; 99:385-91. [PMID: 24056814 DOI: 10.3324/haematol.2013.093062] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We evaluated short-term response endpoints for acute graft-versus-host disease treatment trials. We postulated that response endpoints should correlate with reduced symptom burden and decreased subsequent treatment failure, defined as non-relapse mortality, recurrent malignancy, or additional systemic treatment. The cohort included 303 consecutive patients who received initial systemic steroid treatment for acute graft-versus-host disease. Response was evaluated at day 28 after initial treatment, which in all cases preceded the onset of chronic graft-versus-host disease. At day 28, 36% of patients had a complete response, 26% had a very good partial response, 10% had another degree of partial response (other partial response) and 28% had no response. As expected, the symptom burden was lower in patients with very good partial response compared to those with other partial response. The frequencies of subsequent treatment failure were similar in patients with complete and very good partial responses, but lower than in patients with other partial response or no response at day 28. The frequency of second-line treatment was lower in patients with very good partial response than in those with other partial response. Risk factors associated with a lower probability of complete or very good partial response at day 28 were unrelated or human leukocyte antigen-mismatched related donor grafts and liver or gastrointestinal involvement at onset of initial treatment. Taken together, these results suggest that endpoints in acute graft-versus-host disease treatment trials should distinguish between very good partial response and other partial response. Our results support the use of complete or very good partial response at day 28 as an appropriate short-term primary endpoint.
Collapse
|
134
|
Multiparameter single-cell profiling of human CD4+FOXP3+ regulatory T-cell populations in homeostatic conditions and during graft-versus-host disease. Blood 2013; 122:1802-12. [DOI: 10.1182/blood-2013-02-482539] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Key Points
Single-cell heterogeneity, rather than lineage reprogramming, explains the remarkable complexity and functional diversity of human Tregs. Altered homeostasis of Treg subpopulations in patients developing acute graft-versus-host disease.
Collapse
|
135
|
Rezende BM, Bernardes PTT, Resende CB, Arantes RME, Souza DG, Braga FC, Castor MGM, Teixeira MM, Pinho V. Lithothamnion muelleri controls inflammatory responses, target organ injury and lethality associated with graft-versus-host disease in mice. Mar Drugs 2013; 11:2595-615. [PMID: 23873335 PMCID: PMC3736440 DOI: 10.3390/md11072595] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 12/31/2022] Open
Abstract
Lithothamnion muelleri (Hapalidiaceae) is a marine red alga, which is a member of a group of algae with anti-inflammatory, antitumor, and immunomodulatory properties. The present study evaluated the effects of treatment with Lithothamnion muelleri extract (LM) in a model of acute graft-versus-host disease (GVHD), using a model of adoptive splenocyte transfer from C57BL/6 donors into B6D2F1 recipient mice. Mice treated with LM showed reduced clinical signs of disease and mortality when compared with untreated mice. LM-treated mice had reduced tissue injury, less bacterial translocation, and decreased levels of proinflammatory cytokines and chemokines (interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-C motif) ligand 3 (CCL3) and chemokine (C-C motif) ligand 5 (CCL5)). The polysaccharide-rich fraction derived from LM could inhibit leukocyte rolling and adhesion in intestinal venules, as assessed by intravital microscopy. LM treatment did not impair the beneficial effects of graft-versus-leukaemia (GVL). Altogether, our studies suggest that treatment with Lithothamnion muelleri has a potential therapeutic application in GVHD treatment.
Collapse
Affiliation(s)
- Barbara M. Rezende
- Laboratory of Resolution of Inflammatory Response, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mails: (B.M.R.); (P.T.T.B.); (C.B.R.); (M.G.M.C.)
| | - Priscila T. T. Bernardes
- Laboratory of Resolution of Inflammatory Response, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mails: (B.M.R.); (P.T.T.B.); (C.B.R.); (M.G.M.C.)
| | - Carolina B. Resende
- Laboratory of Resolution of Inflammatory Response, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mails: (B.M.R.); (P.T.T.B.); (C.B.R.); (M.G.M.C.)
| | - Rosa M. E. Arantes
- Laboratory of Experimental Neuro-Immunopathology, Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mail:
| | - Danielle G. Souza
- Host-Microbes Interaction Laboratory, Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mail:
| | - Fernão C. Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mail:
| | - Marina G. M. Castor
- Laboratory of Resolution of Inflammatory Response, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mails: (B.M.R.); (P.T.T.B.); (C.B.R.); (M.G.M.C.)
| | - Mauro M. Teixeira
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mail:
| | - Vanessa Pinho
- Laboratory of Resolution of Inflammatory Response, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil; E-Mails: (B.M.R.); (P.T.T.B.); (C.B.R.); (M.G.M.C.)
| |
Collapse
|
136
|
Inhibiting retinoic acid signaling ameliorates graft-versus-host disease by modifying T-cell differentiation and intestinal migration. Blood 2013; 122:2125-34. [PMID: 23814022 DOI: 10.1182/blood-2012-11-470252] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a critical complication after allogeneic bone marrow transplantation. During GVHD, donor T cells are activated by host antigen-presenting cells and differentiate into T-effector cells (Teffs) that migrate to GVHD target organs. However, local environmental factors influencing Teff differentiation and migration are largely unknown. Vitamin A metabolism within the intestine produces retinoic acid, which contributes to intestinal homeostasis and tolerance induction. Here, we show that the expression and function of vitamin A-metabolizing enzymes were increased in the intestine and mesenteric lymph nodes in mice with active GVHD. Moreover, transgenic donor T cells expressing a retinoic acid receptor (RAR) response element luciferase reporter responded to increased vitamin A metabolites in GVHD-affected organs. Increasing RAR signaling accelerated GVHD lethality, whereas donor T cells expressing a dominant-negative RARα (dnRARα) showed markedly diminished lethality. The dnRARα transgenic T cells showed reduced Th1 differentiation and α4β7 and CCR9 expression associated with poor intestinal migration, low GVHD pathology, and reduced intestinal permeability, primarily via CD4(+) T cells. The inhibition of RAR signaling augmented donor-induced Treg generation and expansion in vivo, while preserving graft-versus-leukemia effects. Together, these results suggested that reagents blunting donor T-cell RAR signaling may possess therapeutic anti-GVHD properties.
Collapse
|
137
|
Israeli M, Klein T, Herscovici C, Ram R, Shpilberg O, Sredni B, Yeshurun M. Cellular immune function monitoring after allogeneic haematopoietic cell transplantation: evaluation of a new assay. Clin Exp Immunol 2013; 172:475-82. [PMID: 23600836 DOI: 10.1111/cei.12072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2013] [Indexed: 02/06/2023] Open
Abstract
Managing the patient's immune system after haematopoietic cell transplantation (HCT) is a challenge, mainly in the unstable period immediately after the transplant. Currently there is no standardized non-invasive diagnostic tool for the evaluation of immunological complications such as graft-versus-host disease (GVHD) and for managing the cellular immune function of the transplant recipient. The ImmuKnow assay for cellular immune function monitoring has been incorporated successfully into the clinical follow-up routine of solid organ transplant recipients. This study aims to explore the relevance and potential contribution of immune monitoring using the assay in the setting of HCT. We found that ImmuKnow-level measurement can distinguish between states of immune function quiescence and between events of acute GVHD. ImmuKnow levels were significantly higher in patients going through GVHD than the levels measured for the same patients during immunological stability. Moreover, we demonstrate a patient case where longitudinal monitoring using the ImmuKnow assay provided a trustworthy depiction of the patient's cellular immune function post-HCT. In conclusion, we provide evidence for the potential contribution of the ImmuKnow assay for longitudinal individualized cellular immune function monitoring of patients following HCT. Further studies are necessary in order to establish the optimal practice for utilizing the assay for this purpose.
Collapse
Affiliation(s)
- M Israeli
- Tissue Typing Laboratory, Rabin Medical Center, Petach-Tikva, Israel.
| | | | | | | | | | | | | |
Collapse
|
138
|
Søndergaard H, Kvist PH, Haase C. Human T cells depend on functional calcineurin, tumour necrosis factor-α and CD80/CD86 for expansion and activation in mice. Clin Exp Immunol 2013; 172:300-10. [PMID: 23574326 DOI: 10.1111/cei.12051] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2012] [Indexed: 12/15/2022] Open
Abstract
Dysregulated T cells are a hallmark of several autoimmune and inflammatory diseases; thus, models to study human T cells in vivo are advantageous, but limited by lacking insight into human T cell functionality in mice. Using non-obese diabetic (NOD), severe combined immunodeficient (SCID) or recombination activating gene-1 (RAG1)(-/-) and interleukin-2 receptor gamma-chain (IL-2Rγ)(-/-) mice reconstituted with human peripheral blood mononuclear cells (PBMCs), we have studied the mechanisms of human T cell expansion and activation in mice. Injection of human PBMCs into mice caused consistent xeno-engraftment with polyclonal expansion and activation of functional human T cells and production of human cytokines. Human T cell expansion coincided with development of a graft-versus-host disease (GVHD)-like condition observed as weight loss, multi-organ immune infiltration and liver damage. CD8(+) T cells alone were sufficient for expansion and required for disease development; in contrast, CD4(+) T cells alone expanded but did not induce acute disease and, rather, exerted regulatory capacity through CD25(+)CD4(+) T cells. Using various anti-inflammatory compounds, we demonstrated that several T cell-activation pathways controlled T cell expansion and disease development, including calcineurin-, tumour necrosis factor-α and co-stimulatory signalling via the CD80/CD86 pathway, indicating the diverse modes of action used by human T cells during expansion and activation in mice as well as the pharmacological relevance of this model. Overall, these data provide insight into the mechanisms used by human T cells during expansion and activation in mice, and we speculate that PBMC-injected mice may be useful to study intrinsic human T cell functions in vivo and to test T cell-targeting compounds.
Collapse
Affiliation(s)
- H Søndergaard
- Department of Immunopharmacology, Novo Nordisk A/S, Måløv, Denmark.
| | | | | |
Collapse
|
139
|
Hannon M, Lechanteur C, Lucas S, Somja J, Seidel L, Belle L, Bruck F, Baudoux E, Giet O, Chantillon AM, Delvenne P, Drion P, Beguin Y, Humblet-Baron S, Baron F. Infusion of clinical-grade enriched regulatory T cells delays experimental xenogeneic graft-versus-host disease. Transfusion 2013; 54:353-63. [PMID: 23772685 DOI: 10.1111/trf.12279] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 04/18/2013] [Accepted: 04/23/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND We investigated the ability of clinical-grade enriched human regulatory T cells (Treg) to attenuate experimental xenogeneic graft-versus-host disease (GVHD) induced by peripheral blood mononuclear cells (PBMNCs; autologous to Treg) infusion in NSG mice, as well as verified their inability to induce xenogeneic GVHD when infused alone. STUDY DESIGN AND METHODS Human Treg were isolated from peripheral blood apheresis products with a cell separation system (CliniMACS, Miltenyi Biotec GmbH) using a two-step procedure (simultaneous CD8 and CD19 depletion followed by CD25-positive selection) in six independent experiments with six different healthy volunteer donors. Sublethally (2.5 Gy) irradiated NSG mice were given 2 × 10(6) cytapheresis (PBMNC) product cells intravenously (IV) without (PBMNC group) or with 1 × 10(6) Treg (PBMNC + Treg group), while other NSG mice received 2 × 10(6) enriched Treg alone (also in IV; Treg group). RESULTS The first five procedures were successful at obtaining a relatively pure Treg population (defined as >50%), while the sixth procedure, due to a technical problem, was not (Treg purity, 42%). Treg cotransfusion significantly delayed death from xenogeneic GVHD in the first five experiments, (p < 0.0001) but not in the sixth experiment. Importantly, none of the mice given enriched Treg alone (Treg group) experienced clinical signs of GVHD, while, interestingly, the CD4+ cells found in these mice 26 days after transplantation were mainly conventional T cells (median CD25+FoxP3+ cells among human CD4+ total cells were only 2.1, 3.1, and 12.2% in spleen, marrow, and blood, respectively). CONCLUSIONS Infusion of clinical-grade enriched Treg delayed the occurrence of xenogeneic GVHD without inducing toxicity in this murine model.
Collapse
Affiliation(s)
- Muriel Hannon
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, Laboratory of Cell and Genetic Therapy, Department of Pathology, Department of Statistics, GIGA-R, Department of Medicine, Division of Hematology, University of Liège, Liège, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; Red Cross Transfusion Center of Liege, Liège, Belgium; Autoimmune Genetics Laboratory, University of Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Martino M, Recchia AG, Gentile M, Morabito L, Vigna E, Cuzzola M, Morabito F. Diagnostic approaches for identifying acute graft-versus-host disease: what comes next? Immunotherapy 2013; 5:553-6. [PMID: 23725276 DOI: 10.2217/imt.13.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
141
|
Conrad DP, Tsang J, Maclean M, Diallo JS, Le Boeuf F, Lemay CG, Falls TJ, Parato KA, Bell JC, Atkins HL. Leukemia cell-rhabdovirus vaccine: personalized immunotherapy for acute lymphoblastic leukemia. Clin Cancer Res 2013; 19:3832-43. [PMID: 23714728 DOI: 10.1158/1078-0432.ccr-12-3199] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Acute lymphoblastic leukemia (ALL) remains incurable in most adults. It has been difficult to provide effective immunotherapy to improve outcomes for the majority of patients. Rhabdoviruses induce strong antiviral immune responses. We hypothesized that mice administered ex vivo rhabdovirus-infected ALL cells [immunotherapy by leukemia-oncotropic virus (iLOV)] would develop robust antileukemic immune responses capable of controlling ALL. EXPERIMENTAL DESIGN Viral protein production, replication, and cytopathy were measured in human and murine ALL cells exposed to attenuated rhabdovirus. Survival following injection of graded amounts of ALL cells was compared between cohorts of mice administered γ-irradiated rhabdovirus-infected ALL cells (iLOV) or multiple control vaccines to determine key immunotherapeutic components and characteristics. Host immune requirements were assessed in immunodeficient and bone marrow-transplanted mice or by adoptive splenocyte transfer from immunized donors. Antileukemic immune memory was ascertained by second leukemic challenge in long-term survivors. RESULTS Human and murine ALL cells were infected and killed by rhabdovirus; this produced a potent antileukemia vaccine. iLOV protected mice from otherwise lethal ALL by developing durable leukemia-specific immune-mediated responses (P < 0.0001), which required an intact CTL compartment. Preexisting antiviral immunity augmented iLOV potency. Splenocytes from iLOV-vaccinated donors protected 60% of naïve recipients from ALL challenge (P = 0.0001). Injecting leukemia cells activated by, or concurrent with, multiple Toll-like receptor agonists could not reproduce the protective effect of iLOV. Similarly, injecting uninfected irradiated viable, apoptotic, or necrotic leukemia cells with/without concurrent rhabdovirus administration was ineffective. CONCLUSION Rhabdovirus-infected leukemia cells can be used to produce a vaccine that induces robust specific immunity against aggressive leukemia.
Collapse
Affiliation(s)
- David P Conrad
- Ottawa Hospital Research Institute, Center for Cancer Therapeutics, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Sellar RS, Peggs KS. Recent progress in managing graft-versus-host disease and viral infections following allogeneic stem cell transplantation. Future Oncol 2013; 8:1549-65. [PMID: 23231517 DOI: 10.2217/fon.12.153] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite recent reductions in transplant-related mortality, post-transplant complications such as graft-versus-host disease (GvHD) remain major obstacles to the successful application of allogeneic hematopoietic transplantation. Steroid-refractory GvHD has a poor outcome. Although there are a variety of new approaches to the treatment of refractory GvHD, many have limited evidence of efficacy. Other approaches appear to be unacceptably toxic. It would be preferable to improve GvHD prophylaxis. There is good evidence that rates of GvHD can be reduced without unacceptable reduction of the graft-versus-leukemia effect or compromising overall survival. However, prophylactic measures aimed at reducing T-cell numbers or functions are associated with high rates of reactivation of latent viruses. New technologies that allow rapid generation of virus-specific T-cells show promise to reduce the frequency and severity of such reactivations and have the potential to revolutionize the approach to post-transplant infectious complications.
Collapse
Affiliation(s)
- Rob S Sellar
- UCL Cancer Institute, Department of Haematology, London, WC1E 6BT, UK
| | | |
Collapse
|
143
|
Holbro A, Lehmann T, Girsberger S, Stern M, Gambazzi F, Lardinois D, Heim D, Passweg JR, Tichelli A, Bubendorf L, Savic S, Hostettler K, Grendelmeier P, Halter JP, Tamm M. Lung histology predicts outcome of bronchiolitis obliterans syndrome after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2013; 19:973-80. [PMID: 23562737 PMCID: PMC7110693 DOI: 10.1016/j.bbmt.2013.03.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 03/25/2013] [Indexed: 12/02/2022]
Abstract
Bronchiolitis obliterans (BO) is a severe complication after allogeneic hematopoietic stem cell transplantation with an unfavorable prognosis. Lung biopsy remains the gold standard for diagnosis. In this retrospective single-center study, we describe 33 patients who underwent biopsy for suspected BO. Ten patients had constrictive BO (CBO); 9 had lymphocytic bronchiolitis (LB), characterized by lymphocytic infiltration of the bronchioles. Six additional patients (4, CBO; 2, LB) had concomitant infection; 8 had other pathological diagnoses. Seven patients with CBO and 3 with LB met the National Institutes of Health consensus BO syndrome definition criteria. An additional 7 patients with histologically confirmed CBO did not meet the consensus definition, 4 of them because of concomitant airway infection. At diagnosis, there were no significant differences between the CBO and LB groups in clinical presentation; pulmonary function tests (median forced expiratory volume in one second [FEV1] at baseline, 90.4% and 99% predicted, at time of video-assisted thoracoscopic surgery, 55.1% and 60.8% for CBO and LB groups, respectively); and chest scans. Treatment was similar in both groups but outcome was different depending on histological findings. FEV1 significantly improved in LB patients compared with CBO patients. Survivals at 1 and 3 years were 77% ± 12% and 60% ± 14% for patients with CBO and 91% ± 9% for patients with LB (P = .028). Lung biopsy in patients with suspected BO enables better characterization of the pattern of BO syndrome. In contrast to CBO, LB is associated with a good long-term prognosis.
Collapse
Affiliation(s)
- Andreas Holbro
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Long-term follow-up of corticosteroid refractory acute GVHD treated with an Inolimomab-based algorithm: a single center experience. Bone Marrow Transplant 2013; 48:1243-8. [DOI: 10.1038/bmt.2013.16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 07/19/2012] [Accepted: 12/29/2012] [Indexed: 12/22/2022]
|
145
|
Graft versus host disease: clinical evaluation, diagnosis and management. Graefes Arch Clin Exp Ophthalmol 2013; 251:1257-66. [PMID: 23504086 DOI: 10.1007/s00417-013-2301-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/23/2013] [Accepted: 02/25/2013] [Indexed: 10/27/2022] Open
Abstract
Graft versus H\host disease (GVHD) can be a devastating complication following bone marrow transplantation. Acute or chronic systemic GVHD can be lethal, and severe damage of different organs and tissues can occur with both types of GVHD. Ocular involvement, either in an acute or chronic presentation, may range from mild to severe with accompanying vision loss present in 60-90 % of patients. Chronic ocular GVHD, the most common form of GVHD, affects mainly the lacrimal gland, meibomian glands, cornea and conjunctiva, mimicking other immunologically mediated inflammatory diseases of the ocular surface without specific symptoms or signs. However, dry eye disease is the main manifestation of GVHD. The long-term treatment of ocular GVHD continues to be challenging and involves a multidisciplinary approach wherein the ophthalmologist plays a major role. Besides systemic immunosuppression and ocular lubricants, topical steroids and topical cyclosporine are commonly prescribed. Newer therapeutic interventions for moderate and severe ocular GVHD include the use of serum eye drops and scleral contact lenses. In this manuscript, we review the mechanisms, clinical findings, and treatment of ocular GVHD.
Collapse
|
146
|
Fong WS, Loh Y, Yoong J. Haematopoietic Stem Cell Transplantation in Systemic Lupus Erythematosus: A Case Report and Review of the Literature. PROCEEDINGS OF SINGAPORE HEALTHCARE 2013. [DOI: 10.1177/201010581302200109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Weng Seng Fong
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore
| | - Yvonne Loh
- Department of Haematology, Singapore General Hospital, Singapore
- Medical Director, Haematopoietic Stem Cell Transplant Program, Singapore General Hospital, Singapore
| | - Jon Yoong
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-NUS Graduate Medical School, Singapore
| |
Collapse
|
147
|
Mochizuki K, Xie F, He S, Tong Q, Liu Y, Mochizuki I, Guo Y, Kato K, Yagita H, Mineishi S, Zhang Y. Delta-like ligand 4 identifies a previously uncharacterized population of inflammatory dendritic cells that plays important roles in eliciting allogeneic T cell responses in mice. THE JOURNAL OF IMMUNOLOGY 2013; 190:3772-82. [PMID: 23440416 DOI: 10.4049/jimmunol.1202820] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Graft-versus-host disease (GVHD) reflects an exaggerated inflammatory allogeneic T cell response in hosts receiving allogeneic hematopoietic stem cell transplantation (HSCT). Inhibition of pan-Notch receptor signaling in donor T cells causes reduction of GVHD. However, which Notch ligand(s) in what APCs is important for priming graft-versus-host reaction remains unknown. We demonstrate that δ-like ligand-4 (Dll4) and Dll4-positive (Dll4(high)) inflammatory dendritic cells (i-DCs) play important roles in eliciting allogeneic T cell responses. Host-type Dll4(high) i-DCs occurred in the spleen and intestine of HSCT mice during GVHD induction phase. These Dll4(high) i-DCs were CD11c(+)B220(+)PDCA-1(+), resembling plasmacytoid dentritic cells (pDCs) of naive mice. However, as compared with unstimulated pDCs, Dll4(high) i-DCs expressed higher levels of costimulatory molecules, Notch ligands Jagged1 and Jagged2, and CD11b, and produced more Ifnb and Il23 but less Il12. In contrast, Dll4-negative (Dll4(low)) i-DCs were CD11c(+)B220(-)PDCA-1(-), and had low levels of Jagged1. In vitro assays showed that Dll4(high) i-DCs induced significantly more IFN-γ- and IL-17-producing effector T cells (3- and 10-fold, respectively) than Dll4(low) i-DCs. This effect could be blocked by anti-Dll4 Ab. In vivo administration of Dll4 Ab reduced donor-alloreactive effector T cells producing IFN-γ and IL-17 in GVHD target organs, leading to reduction of GVHD and improved survival of mice after allogeneic HSCT. Our findings indicate that Dll4(high) i-DCs represent a previously uncharacterized i-DC population distinctive from steady state DCs and Dll4(low) i-DCs. Furthermore, Dll4 and Dll4(high) i-DCs may be beneficial targets for modulating allogeneic T cell responses, and could facilitate the discovery of human counterparts of mouse Dll4(high) i-DCs.
Collapse
Affiliation(s)
- Kazuhiro Mochizuki
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Rowley SD, Friedman T, Korngold R. Hematopoietic stem cell transplantation for malignant diseases. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00097-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
149
|
Wang L, Romero M, Ratajczak P, Lebœuf C, Belhadj S, Peffault de Latour R, Zhao WL, Socié G, Janin A. Increased apoptosis is linked to severe acute GVHD in patients with Fanconi anemia. Bone Marrow Transplant 2012; 48:849-53. [PMID: 23222379 DOI: 10.1038/bmt.2012.237] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Fanconi anemia (FA) patients have an increased risk of acute GVHD (aGVHD) after hematopoietic SCT, with hypersensitivity to DNA-cross-linking agents and defective DNA repair. MicroRNA-34 and p53 can induce apoptosis after DNA damage.Here we assessed epithelial cell apoptosis, and studied TP53 and miR-34a expression in the skin and gut biopsies in five non-transplanted FA patients, in 20 FA patients with aGVHD and in 25 acquired aplastic anemia patients (AA). Epithelial apoptosis was higher in FA than in acquired AA patients in both the skin and gut biopsies, though they had a similar preparative regimen. Further study on gut biopsies in FA patients showed that this deleterious effect was not linked to TP53 gene overexpression. As, among p53-independent signaling pathways of apoptosis, the microRNA-34 family mimics p53 apoptotic effects in response to DNA damage, we studied miR-34a expression in the same series of FA patients' gut biopsies. MiR-34a expression level was higher in severe aGVHD compared with non-aGVHD subjects or non-transplanted patients, and significantly related to apoptotic cell numbers across the three groups of FA patients. Thus, in FA patients, increased apoptosis occurs in target epithelial cells of severe aGVHD, and this deleterious effect is linked to overexpression of miR-34a but not TP53.
Collapse
Affiliation(s)
- L Wang
- Inserm, U728, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
150
|
van der Waart AB, van der Velden WJFM, van Halteren AGS, Leenders MJLG, Feuth T, Blijlevens NMA, van der Voort R, Dolstra H. Decreased levels of circulating IL17-producing CD161+CCR6+ T cells are associated with graft-versus-host disease after allogeneic stem cell transplantation. PLoS One 2012; 7:e50896. [PMID: 23226545 PMCID: PMC3514180 DOI: 10.1371/journal.pone.0050896] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/26/2012] [Indexed: 12/23/2022] Open
Abstract
The C-type lectin-like receptor CD161 is a well-established marker for human IL17-producing T cells, which have been implicated to contribute to the development of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (allo-SCT). In this study, we analyzed CD161+ T cell recovery, their functional properties and association with GVHD occurrence in allo-SCT recipients. While CD161+CD4+ T cells steadily recovered, CD161hiCD8+ T cell numbers declined during tapering of Cyclosporine A (CsA), which can be explained by their initial growth advantage over CD161neg/lowCD8+ T cells due to ABCB1-mediated CsA efflux. Interestingly, occurrence of acute and chronic GVHD was significantly correlated with decreased levels of circulating CD161+CD4+ as well as CD161hiCD8+ T cells. In addition, these subsets from transplanted patients secreted high levels of IFNγ and IL17. Moreover, we found that CCR6 co-expression by CD161+ T cells mediated specific migration towards CCL20, which was expressed in GVHD biopsies. Finally, we demonstrated that CCR6+ T cells indeed were present in these CCL20+ GVHD-affected tissues. In conclusion, we showed that functional CD161+CCR6+ co-expressing T cells disappear from the circulation and home to GVHD-affected tissue sites. These findings support the hypothesis that CCR6+CD161-expressing T cells may be involved in the immune pathology of GVHD following their CCL20-dependent recruitment into affected tissues.
Collapse
Affiliation(s)
- Anniek B. van der Waart
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | - Astrid G. S. van Halteren
- Immunology Laboratory, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marij J. L. G. Leenders
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Ton Feuth
- Department of Epidemiology, Biostatistics and Health Technology Assessment, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Nicole M. A. Blijlevens
- Department of Hematology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Robbert van der Voort
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|