1
|
McDaniel Mims B, Furr KL, Enriquez J, Grisham MB. Improving bench-to-bedside translation for acute graft-versus-host disease models. Dis Model Mech 2025; 18:DMM052084. [PMID: 40019007 PMCID: PMC11892683 DOI: 10.1242/dmm.052084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025] Open
Abstract
The transplantation of allogeneic hematopoietic stem cells is a potentially curative treatment for hematological malignancies, inherited blood disorders and immune deficiencies. Unfortunately, 30-50% of patients receiving allogeneic hematopoietic stem cells will develop a potentially life-threatening inflammatory disease called acute graft-versus-host disease (aGVHD). In patients with aGVHD, graft-associated T cells, which typically target the skin, intestinal tract and liver, can also damage the lungs and lymphoid tissue. Damage to lymphoid tissue creates prolonged immunodeficiency that markedly increases the risk of infections and bleeding, resulting in considerable morbidity and mortality. Although mouse models of aGVHD have been instrumental to our understanding of this condition's pathogenesis, translation of preclinical data into new and more effective treatments for human disease has been limited for reasons that remain to be fully understood. However, evidence suggests that factors associated with mouse models of aGVHD likely contribute to these unsatisfactory results. In this Review, we identify and discuss the specific factors inherent to mouse models of aGVHD that may limit the translation of preclinical data to patient treatment, and suggest how to improve the translatability of these models.
Collapse
Affiliation(s)
- Brianyell McDaniel Mims
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| | - Josue Enriquez
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| |
Collapse
|
2
|
Jansen SA, Cutilli A, de Koning C, van Hoesel M, Frederiks CL, Saiz Sierra L, Nierkens S, Mokry M, Nieuwenhuis EE, Hanash AM, Mocholi E, Coffer PJ, Lindemans CA. Chemotherapy-induced intestinal epithelial damage directly promotes galectin-9-driven modulation of T cell behavior. iScience 2024; 27:110072. [PMID: 38883813 PMCID: PMC11176658 DOI: 10.1016/j.isci.2024.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/05/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
The intestine is vulnerable to chemotherapy-induced damage due to the high rate of intestinal epithelial cell (IEC) proliferation. We have developed a human intestinal organoid-based 3D model system to study the direct effect of chemotherapy-induced IEC damage on T cell behavior. Exposure of intestinal organoids to busulfan, fludarabine, and clofarabine induced damage-related responses affecting both the capacity to regenerate and transcriptional reprogramming. In ex vivo co-culture assays, prior intestinal organoid damage resulted in increased T cell activation, proliferation, and migration. We identified galectin-9 (Gal-9) as a key molecule released by damaged organoids. The use of anti-Gal-9 blocking antibodies or CRISPR/Cas9-mediated Gal-9 knock-out prevented intestinal organoid damage-induced T cell proliferation, interferon-gamma release, and migration. Increased levels of Gal-9 were found early after HSCT chemotherapeutic conditioning in the plasma of patients who later developed acute GVHD. Taken together, chemotherapy-induced intestinal damage can influence T cell behavior in a Gal-9-dependent manner which may provide novel strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Suze A. Jansen
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Alessandro Cutilli
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Coco de Koning
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584GX Utrecht, the Netherlands
| | - Marliek van Hoesel
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Cynthia L. Frederiks
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Leire Saiz Sierra
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584GX Utrecht, the Netherlands
| | - Michal Mokry
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
| | - Edward E.S. Nieuwenhuis
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- University College Roosevelt, Utrecht University, Middelburg 4331CB, the Netherlands
| | - Alan M. Hanash
- Departments of Medicine and Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY 10065, USA
| | - Enric Mocholi
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Paul J. Coffer
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Caroline A. Lindemans
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| |
Collapse
|
3
|
Schreiber F, Balas I, Robinson MJ, Bakdash G. Border Control: The Role of the Microbiome in Regulating Epithelial Barrier Function. Cells 2024; 13:477. [PMID: 38534321 PMCID: PMC10969408 DOI: 10.3390/cells13060477] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/28/2024] Open
Abstract
The gut mucosal epithelium is one of the largest organs in the body and plays a critical role in regulating the crosstalk between the resident microbiome and the host. To this effect, the tight control of what is permitted through this barrier is of high importance. There should be restricted passage of harmful microorganisms and antigens while at the same time allowing the absorption of nutrients and water. An increased gut permeability, or "leaky gut", has been associated with a variety of diseases ranging from infections, metabolic diseases, and inflammatory and autoimmune diseases to neurological conditions. Several factors can affect gut permeability, including cytokines, dietary components, and the gut microbiome. Here, we discuss how the gut microbiome impacts the permeability of the gut epithelial barrier and how this can be harnessed for therapeutic purposes.
Collapse
Affiliation(s)
| | | | | | - Ghaith Bakdash
- Microbiotica Ltd., Cambridge CB10 1XL, UK; (F.S.); (I.B.); (M.J.R.)
| |
Collapse
|
4
|
Wang YM, Abdullah S, Luebbering N, Langenberg L, Duell A, Lake K, Lane A, Hils B, Vazquez Silva O, Trapp M, Nalapareddy K, Koo J, Denson LA, Jodele S, Haslam DB, Faubion WA, Davies SM, Khandelwal P. Intestinal permeability in patients undergoing stem cell transplantation correlates with systemic acute phase responses and dysbiosis. Blood Adv 2023; 7:5137-5151. [PMID: 37083597 PMCID: PMC10480541 DOI: 10.1182/bloodadvances.2023009960] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
Intestinal permeability may correlate with adverse outcomes during hematopoietic stem cell transplantation (HSCT), but longitudinal quantification with traditional oral mannitol and lactulose is not feasible in HSCT recipients because of mucositis and diarrhea. A modified lactulose:rhamnose (LR) assay is validated in children with environmental enteritis. Our study objective was to quantify peri-HSCT intestinal permeability changes using the modified LR assay. The LR assay was administered before transplant, at day +7 and +30 to 80 pediatric and young adult patients who received allogeneic HSCT. Lactulose and rhamnose were detected using urine mass spectrometry and expressed as an L:R ratio. Metagenomic shotgun sequencing of stool for microbiome analyses and enzyme-linked immunosorbent assay analyses of plasma lipopolysaccharide binding protein (LBP), ST2, REG3α, claudin1, occludin, and intestinal alkaline phosphatase were performed at the same timepoints. L:R ratios were increased at day +7 but returned to baseline at day +30 in most patients (P = .014). Conditioning regimen intensity did not affect the trajectory of L:R (P = .39). Baseline L:R ratios did not vary with diagnosis. L:R correlated with LBP levels (r2 = 0.208; P = .0014). High L:R ratios were associated with lower microbiome diversity (P = .035), loss of anaerobic organisms (P = .020), and higher plasma LBP (P = .0014). No adverse gastrointestinal effects occurred because of LR. Intestinal permeability as measured through L:R ratios after allogeneic HSCT correlates with intestinal dysbiosis and elevated plasma LBP. The LR assay is well-tolerated and may identify transplant recipients who are more likely to experience adverse outcomes.
Collapse
Affiliation(s)
- YunZu Michele Wang
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Sheyar Abdullah
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Nathan Luebbering
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Lucille Langenberg
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Alexandra Duell
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kelly Lake
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Brian Hils
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Ormarie Vazquez Silva
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Monica Trapp
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kodandaramireddy Nalapareddy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jane Koo
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Lee A. Denson
- University of Cincinnati College of Medicine, Cincinnati, OH
- Department of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - David B. Haslam
- University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | | | - Stella M. Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Pooja Khandelwal
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
5
|
Jansen SA, Cutilli A, de Koning C, van Hoesel M, Sierra LS, Nierkens S, Mokry M, Nieuwenhuis EES, Hanash AM, Mocholi E, Coffer PJ, Lindemans CA. Chemotherapy-induced intestinal injury promotes Galectin-9-driven modulation of T cell function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.30.538862. [PMID: 37163028 PMCID: PMC10168344 DOI: 10.1101/2023.04.30.538862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The intestine is vulnerable to chemotherapy-induced toxicity due to its high epithelial proliferative rate, making gut toxicity an off-target effect in several cancer treatments, including conditioning regimens for allogeneic hematopoietic cell transplantation (allo-HCT). In allo-HCT, intestinal damage is an important factor in the development of Graft-versus-Host Disease (GVHD), an immune complication in which donor immune cells attack the recipient's tissues. Here, we developed a novel human intestinal organoid-based 3D model system to study the direct effect of chemotherapy-induced intestinal epithelial damage on T cell behavior. Chemotherapy treatment using busulfan, fludarabine, and clofarabine led to damage responses in organoids resulting in increased T cell migration, activation, and proliferation in ex- vivo co-culture assays. We identified galectin-9 (Gal-9), a beta-galactoside-binding lectin released by damaged organoids, as a key molecule mediating T cell responses to damage. Increased levels of Gal-9 were also found in the plasma of allo-HCT patients who later developed acute GVHD, supporting the predictive value of the model system in the clinical setting. This study highlights the potential contribution of chemotherapy-induced epithelial damage to the pathogenesis of intestinal GVHD through direct effects on T cell activation and trafficking promoted by galectin-9.
Collapse
|
6
|
Horowitz A, Chanez-Paredes SD, Haest X, Turner JR. Paracellular permeability and tight junction regulation in gut health and disease. Nat Rev Gastroenterol Hepatol 2023:10.1038/s41575-023-00766-3. [PMID: 37186118 PMCID: PMC10127193 DOI: 10.1038/s41575-023-00766-3] [Citation(s) in RCA: 265] [Impact Index Per Article: 132.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2023] [Indexed: 05/17/2023]
Abstract
Epithelial tight junctions define the paracellular permeability of the intestinal barrier. Molecules can cross the tight junctions via two distinct size-selective and charge-selective paracellular pathways: the pore pathway and the leak pathway. These can be distinguished by their selectivities and differential regulation by immune cells. However, permeability increases measured in most studies are secondary to epithelial damage, which allows non-selective flux via the unrestricted pathway. Restoration of increased unrestricted pathway permeability requires mucosal healing. By contrast, tight junction barrier loss can be reversed by targeted interventions. Specific approaches are needed to restore pore pathway or leak pathway permeability increases. Recent studies have used preclinical disease models to demonstrate the potential of pore pathway or leak pathway barrier restoration in disease. In this Review, we focus on the two paracellular flux pathways that are dependent on the tight junction. We discuss the latest evidence that highlights tight junction components, structures and regulatory mechanisms, their impact on gut health and disease, and opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Arie Horowitz
- UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, Normandie University, Rouen, France
| | - Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Haroun E, Kumar PA, Saba L, Kassab J, Ghimire K, Dutta D, Lim SH. Intestinal barrier functions in hematologic and oncologic diseases. J Transl Med 2023; 21:233. [PMID: 37004099 PMCID: PMC10064590 DOI: 10.1186/s12967-023-04091-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023] Open
Abstract
The intestinal barrier is a complex structure that not only regulates the influx of luminal contents into the systemic circulation but is also involved in immune, microbial, and metabolic homeostasis. Evidence implicating disruption in intestinal barrier functions in the development of many systemic diseases, ranging from non-alcoholic steatohepatitis to autism, or systemic complications of intestinal disorders has increased rapidly in recent years, raising the possibility of the intestinal barrier as a potential target for therapeutic intervention to alter the course and mitigate the complications associated with these diseases. In addition to the disease process being associated with a breach in the intestinal barrier functions, patients with hematologic and oncologic diseases are particularly at high risks for the development of increased intestinal permeability, due to the frequent use of broad-spectrum antibiotics and chemoradiation. They also face a distinct challenge of being intermittently severely neutropenic due to treatment of the underlying conditions. In this review, we will discuss how hematologic and oncologic diseases are associated with disruption in the intestinal barrier and highlight the complications associated with an increase in the intestinal permeability. We will explore methods to modulate the complication. To provide a background for our discussion, we will first examine the structure and appraise the methods of evaluation of the intestinal barrier.
Collapse
Affiliation(s)
- Elio Haroun
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA
| | - Prashanth Ashok Kumar
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA
| | - Ludovic Saba
- Department of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Joseph Kassab
- Department of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Krishna Ghimire
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA
| | - Dibyendu Dutta
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA.
| | - Seah H Lim
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA.
| |
Collapse
|
8
|
Oikonomopoulou C, Paisiou A, Ioannidou ED, Komitopoulou A, Kaisari A, Zisaki K, Kastamoulas M, Stavroulaki G, Giannakopoulou A, Vessalas G, Kitra-Roussou V, Goussetis E, Peristeri I. Allogeneic hematopoietic stem cell transplantation in infants is associated with significant morbidity and mortality. Pediatr Transplant 2022; 26:e14239. [PMID: 35122456 DOI: 10.1111/petr.14239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Infants are subjected to hematopoietic stem cell transplantation (HSCT) due to malignant and non-malignant diseases. However, specific data concerning the outcome and transplantation-related complications in infants, as a separate age group, are limited. Our aim was to evaluate the impact of infancy on the outcome, toxicity, and complications after HSCT. METHODS We retrospectively analyzed data of 55 infants that underwent HSCT in our unit from May 1997 until February 2020, emphasizing on the probability of overall survival (OS) and the cumulative incidence (CI) of transplantation-related mortality (TRM) and complications. RESULTS We report a probability of OS of 61%, a CI of TRM at day 100 and 365 post transplantation of 22% and 30%, respectively, and additionally a CI of graft failure, acute graft-versus-host disease (GvHD), and infectious complications, 18%, 44%, and 39%, respectively. No statistically significant association was detected between the above mentioned parameters and diagnosis, the use of myeloablative or non-myeloablative/reduced toxicity conditioning regimens or the type of donor. CONCLUSIONS We conclude that HSCT in infancy is associated with significant mortality and morbidity. This is possibly attributed to endogenous, age-related factors. More specifically, infants may be at a higher risk of toxicities due to the immaturity of developing vital organs and the deficiency of the newly adopted immune system that predisposes them to infectious complications. The development of GvHD further augments the danger of infections, in a potential vice-versa relationship. Moreover, there are few data on pharmacokinetics of chemotherapy agents, making safe and efficacious drug administration hard.
Collapse
Affiliation(s)
| | - Anna Paisiou
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| | | | - Anna Komitopoulou
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| | - Aikaterini Kaisari
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| | - Kalliopi Zisaki
- Transfusion Department, Agia Sofia Children's Hospital, Athens, Greece
| | | | | | - Aikaterini Giannakopoulou
- First Department of Paediatrics, National and Kapodistrian University of Athens, Agia Sophia Children's Hospital, Athens, Greece
| | - George Vessalas
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| | | | - Evgenios Goussetis
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| | - Ioulia Peristeri
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| |
Collapse
|
9
|
Challenges and opportunities targeting mechanisms of epithelial injury and recovery in acute intestinal graft-versus-host disease. Mucosal Immunol 2022; 15:605-619. [PMID: 35654837 PMCID: PMC9259481 DOI: 10.1038/s41385-022-00527-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023]
Abstract
Despite advances in immunosuppressive prophylaxis and overall supportive care, gastrointestinal (GI) graft-versus-host disease (GVHD) remains a major, lethal side effect after allogeneic hematopoietic stem cell transplantation (allo-HSCT). It has become increasingly clear that the intestinal epithelium, in addition to being a target of transplant-related toxicity and GVHD, plays an important role in the onset of GVHD. Over the last two decades, increased understanding of the epithelial constituents and their microenvironment has led to the development of novel prophylactic and therapeutic interventions, with the potential to protect the intestinal epithelium from GVHD-associated damage and promote its recovery following insult. In this review, we will discuss intestinal epithelial injury and the role of the intestinal epithelium in GVHD pathogenesis. In addition, we will highlight possible approaches to protect the GI tract from damage posttransplant and to stimulate epithelial regeneration, in order to promote intestinal recovery. Combined treatment modalities integrating immunomodulation, epithelial protection, and induction of regeneration may hold the key to unlocking mucosal recovery and optimizing therapy for acute intestinal GVHD.
Collapse
|
10
|
Haring E, Zeiser R, Apostolova P. Interfering With Inflammation: Heterogeneous Effects of Interferons in Graft- Versus-Host Disease of the Gastrointestinal Tract and Inflammatory Bowel Disease. Front Immunol 2021; 12:705342. [PMID: 34249014 PMCID: PMC8264264 DOI: 10.3389/fimmu.2021.705342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
The intestine can be the target of several immunologically mediated diseases, including graft-versus-host disease (GVHD) and inflammatory bowel disease (IBD). GVHD is a life-threatening complication that occurs after allogeneic hematopoietic stem cell transplantation. Involvement of the gastrointestinal tract is associated with a particularly high mortality. GVHD development starts with the recognition of allo-antigens in the recipient by the donor immune system, which elicits immune-mediated damage of otherwise healthy tissues. IBD describes a group of immunologically mediated chronic inflammatory diseases of the intestine. Several aspects, including genetic predisposition and immune dysregulation, are responsible for the development of IBD, with Crohn’s disease and ulcerative colitis being the two most common variants. GVHD and IBD share multiple key features of their onset and development, including intestinal tissue damage and loss of intestinal barrier function. A further common feature in the pathophysiology of both diseases is the involvement of cytokines such as type I and II interferons (IFNs), amongst others. IFNs are a family of protein mediators produced as a part of the inflammatory response, typically to pathogens or malignant cells. Diverse, and partially paradoxical, effects have been described for IFNs in GVHD and IBD. This review summarizes current knowledge on the role of type I, II and III IFNs, including basic concepts and controversies about their functions in the context of GVHD and IBD. In addition, therapeutic options, research developments and remaining open questions are addressed.
Collapse
Affiliation(s)
- Eileen Haring
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petya Apostolova
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Advances in Intestinal Barrier Preservation and Restoration in the Allogeneic Hematopoietic Cell Transplantation Setting. J Clin Med 2021; 10:jcm10112508. [PMID: 34204044 PMCID: PMC8201017 DOI: 10.3390/jcm10112508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
The intestinal barrier consists of an epithelial lining covered with specialized mucus inhabited by intestinal microbiota. An intact gut barrier ensures a resistance to bacteria and toxins translocation. On the other hand, gut permeability allows the absorption of essential nutrients, fluids and ions. This balance is achieved only by the complex structure and functional characteristics of the intestinal barrier. Allogenic hematopoietic cell transplantation remains the only curative treatment for many hematological diseases, but its application is limited because of possible transplant-related mortality mainly due to graft-versus-host disease and infectious complications. The intestinal barrier has been extensively studied in recent years as the primary site of graft-versus-host disease initiation and propagation. In the present review, we focused on the physiological structure and function of the gut barrier and the evidence of how the disruption of the gut barrier and increased intestinal permeability affects transplant recipients. Finally, therapeutic strategies aiming at intestinal barrier protection with a special focus on microbiome preservation and restoration in the allogenic hematopoietic cell transplantation setting are discussed.
Collapse
|
12
|
Curative potential of fludarabine, melphalan, and non-myeloablative dosage of busulfan in elderly patients with myeloid malignancy. Int J Hematol 2019; 111:247-255. [DOI: 10.1007/s12185-019-02763-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 01/24/2023]
|
13
|
Dorshow RB, Johnson JR, Debreczeny MP, Riley IR, Shieh JJ, Rogers TE, Hall-Moore C, Shaikh N, Rouggly-Nickless LC, Tarr PI. Transdermal fluorescence detection of a dual fluorophore system for noninvasive point-of-care gastrointestinal permeability measurement. BIOMEDICAL OPTICS EXPRESS 2019; 10:5103-5116. [PMID: 31646033 PMCID: PMC6788606 DOI: 10.1364/boe.10.005103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/28/2019] [Accepted: 09/09/2019] [Indexed: 05/10/2023]
Abstract
The intestinal mucosal barrier prevents macromolecules and pathogens from entering the circulatory stream. Tight junctions in this barrier are compromised in inflammatory bowel diseases, environmental enteropathy, and enteric dysfunction. Dual sugar absorption tests are a standard method for measuring gastrointestinal integrity, however, these are not clinically amenable. Herein, we report on a dual fluorophore system and fluorescence detection instrumentation for which gastrointestinal permeability is determined in a rat small bowel disease model from the longitudinal measured transdermal fluorescence of each fluorophore. This fluorophore technology enables a specimen-free, noninvasive, point-of-care gastrointestinal permeability measurement which should be translatable to human clinical studies.
Collapse
Affiliation(s)
| | - J. R. Johnson
- MediBeacon Inc., 1100 Corporate Square Drive, St. Louis, MO 63132, USA
| | | | - I. Rochelle Riley
- MediBeacon Inc., 1100 Corporate Square Drive, St. Louis, MO 63132, USA
| | - Jeng-Jong Shieh
- MediBeacon Inc., 1100 Corporate Square Drive, St. Louis, MO 63132, USA
| | - Thomas E. Rogers
- MediBeacon Inc., 1100 Corporate Square Drive, St. Louis, MO 63132, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Phillip I. Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
14
|
Nalle SC, Zuo L, Ong MLDM, Singh G, Worthylake AM, Choi W, Manresa MC, Southworth AP, Edelblum KL, Baker GJ, Joseph NE, Savage PA, Turner JR. Graft-versus-host disease propagation depends on increased intestinal epithelial tight junction permeability. J Clin Invest 2019; 129:902-914. [PMID: 30667372 PMCID: PMC6355225 DOI: 10.1172/jci98554] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a complication of hematopoietic stem cell transplantation (HSCT) that affects multiple organs. GVHD-associated intestinal damage can be separated into two distinct phases, initiation and propagation, which correspond to conditioning-induced damage and effector T cell activation and infiltration, respectively. Substantial evidence indicates that intestinal damage induced by pretransplant conditioning is a key driver of GVHD initiation. Here, we aimed to determine the impact of dysregulated intestinal permeability on the subsequent GVHD propagation phase. The initiation phase of GVHD was unchanged in mice lacking long MLCK (MLCK210), an established regulator of epithelial tight junction permeability. However, MLCK210-deficient mice were protected from sustained barrier loss and exhibited limited GVHD propagation, as indicated by reduced histopathology, fewer CD8+ effector T cells in the gut, and improved overall survival. Consistent with these findings, intestinal epithelial MLCK210 expression and enzymatic activity were similarly increased in human and mouse GVHD biopsies. Intestinal epithelial barrier loss mediated by MLCK210 is therefore a key driver of the GVHD propagation. These data suggest that inhibition of MLCK210-dependent barrier regulation may be an effective approach to limiting GVHD progression.
Collapse
Affiliation(s)
- Sam C. Nalle
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Li Zuo
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Anhui Medical University, Hefei, Anhui, China
| | - Ma. Lora Drizella M. Ong
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gurminder Singh
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alicia M. Worthylake
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wangsun Choi
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mario Cabrero Manresa
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anna P. Southworth
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Karen L. Edelblum
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology & Laboratory Medicine, Center for Inflammation and Immunity, Rutgers New Jersey Medical School, Cancer Center, Newark, New Jersey, USA
| | - Gregory J. Baker
- Laboratory of Systems Pharmacology, Harvard Medical School, Harvard Program in Therapeutic Science, Boston, Massachusetts, USA
| | - Nora E. Joseph
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Peter A. Savage
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Jerrold R. Turner
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Early Blood Stream Infections after BMT are Associated with Cytokine Dysregulation and Poor Overall Survival. Biol Blood Marrow Transplant 2018. [DOI: 10.1016/j.bbmt.2018.02.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Association Between Low Plasma Level of Citrulline Before Allogeneic Hematopoietic Cell Transplantation and Severe Gastrointestinal Graft vs Host Disease. Clin Gastroenterol Hepatol 2018; 16:908-917.e2. [PMID: 29258901 DOI: 10.1016/j.cgh.2017.12.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/06/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The gastrointestinal form of acute graft vs host disease increases morbidity and mortality following allogeneic hematopoietic cell transplantation. Plasma levels of citrulline, a non-proteinogenic amino acid, indicate functional enterocyte mass. We measured citrulline in patients before allogeneic hematopoietic cell transplantation and investigated its association with incidence and severity of gastrointestinal graft vs host disease. METHODS We performed a retrospective study with 191 patients (69 women, 122 men; median age of 52 years) who underwent allogeneic hematopoietic cell transplantation for hematological malignancies at a tertiary center of France from January 2013 through April 2015. Levels of citrulline in plasma samples collected 30 days before graft infusion were measured by high performance liquid chromatography with tandem mass spectrometry. We assigned patients to groups with a high level of citrulline (>26 μmol/L) or low level of citrulline (≤26 μmol/L). The primary outcomes were difference between groups in incidence of stage 2-4 gastrointestinal graft vs host disease, death without hematological disease relapse (non-relapse mortality), relapse of the hematological disease, and overall survival through 2 years after transplantation. RESULTS Ninety-six patients (50%) developed acute graft vs host disease and 37 (19%) developed a gastrointestinal form. Among patients with gastrointestinal involvement, 33 patients (89%) had stage 2-4 gastrointestinal graft vs host disease. In univariable analysis, low level of citrulline associated with higher cumulative incidence of stage 2-4 gastrointestinal graft vs host disease, non-relapse mortality, and shorter overall survival. In multivariable analysis, low level of citrulline was the only risk factor independently associated with stage 2-4 gastrointestinal graft vs host disease (hazard ratio, 3.06; 95% CI, 1.37-6.85; P = .007); it also associated with increased non-relapse mortality (hazard ratio, 2.29; 95% CI, 1.24-4.22; P = .008). CONCLUSIONS In a retrospective study with 191 patients, we associated a low plasma level of citrulline before allogeneic hematopoietic cell transplantation with a higher risk for stage 2-4 gastrointestinal graft vs host disease and non-relapse mortality. This marker might be used to manage patients before allogeneic hematopoietic cell transplantation.
Collapse
|
17
|
da Costa LNG, Costa-Lima C, de Meirelles LR, Carvalho RB, Colella MP, Aranha FJP, Vigorito AC, De Paula EV. Association between histopathological alterations and diarrhea severity in acute intestinal graft-versus-host disease. Medicine (Baltimore) 2018; 97:e0600. [PMID: 29742694 PMCID: PMC5959433 DOI: 10.1097/md.0000000000010600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gastrointestinal (GI) acute graft-versus-host disease (aGVHD) remains one of the most important complications of allogeneic hematopoietic cell transplantation (allo-HCT). The diagnosis of this complication is largely dependent on clinical symptoms, but GI biopsies are warranted in most cases, due to the multitude of potential causes that coexist in patients with a clinical suspicion of this complication. In addition, several lines of evidence support that the GI is not only a target organ in aGVHD, but also a key mediator of the pathogenesis of this condition. Controversy exists on whether histopathological findings are associated with clinical severity. Crypt loss is a relatively straightforward histological finding of GI aGVHD, whose presence has been associated with disease severity in a previous study.In order to independently validate this association, we retrospectively evaluated all histological changes from 25 patients with confirmed GI aGVHD who underwent allo-HCT in our center from 2008 to 2014. Clinical, laboratory, and histological data were obtained from the medical records and pathological reports. All GI biopsies were reviewed by 2 investigators blinded to clinical data, who classified GI aGVHD according to the presence of severe crypt loss.The proportion of patients with grades I-II and III-IV aGVHD patients in our population were 45.5% and 54.5%, respectively. The most common histological alterations were isolated apoptotic bodies, present in 80% of colon biopsies with aGVHD. Severe crypt loss, corresponding to grades III-IV aGVHD was associated with higher stool volumes (P = .02) and increased diarrhea duration (P = .02), but not with response to steroids or mortality.In this study, we independently validated that the presence of severe crypt loss, a reliable and simple parameter to grade the extension of GI aGVHD, is associated with disease severity in GI aGVHD.
Collapse
Affiliation(s)
| | - Carolina Costa-Lima
- Faculty of Medical Sciences
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP
| | | | | | | | | | | | - Erich Vinicius De Paula
- Faculty of Medical Sciences
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP
| |
Collapse
|
18
|
Sano H, Hilinski JA, Qayed M, Applegate K, Newton JG, Watkins B, Chiang KY, Horan J. Early blood stream infection following allogeneic hematopoietic stem cell transplantation is a risk factor for acute grade III-IV GVHD in children and adolescents. Pediatr Blood Cancer 2018; 65. [PMID: 28921804 DOI: 10.1002/pbc.26821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Graft-versus-host disease (GVHD) remains a major cause of mortality and morbidity in allogeneic hematopoietic stem cell transplantation (HSCT). In adults, early blood stream infection (BSI) and acute GVHD (AGVHD) have been reported to be related. The impact of BSI on risk for AGVHD, however, has not been assessed in pediatric patients. PROCEDURE We conducted a retrospective analysis to test the hypothesis that early BSI (before day +30) predisposes allogeneic pediatric transplant patients to severe AGVHD. We analyzed 293 allogeneic HSCT performed at Children's Healthcare of Atlanta between 2005 and 2014 that met eligibility criteria. RESULTS The cumulative incidence of acute grade III-IV GVHD at 100 days after HSCT was 17.1%. In multivariate analysis, risk for acute grade III-IV GVHD was associated with HLA-mismatched donor (hazard ratio [HR] = 4.870, P < 0.001), and BSI between day 0 and +30 prior to AGVHD (HR = 3.010, P = 0.001). CONCLUSIONS These results indicate that early BSI appears to be a risk factor for acute grade III-IV GVHD. Further research is needed to determine if the link is causal.
Collapse
Affiliation(s)
- Hirozumi Sano
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Joseph A Hilinski
- Division of Pediatric Infectious Diseases, St. Luke's Children's Hospital, Boise, Idaho
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Kristy Applegate
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Joanna G Newton
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Benjamin Watkins
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Kuang-Yueh Chiang
- Department of Pediatrics, Haematology/Oncology, the Hospital for Sick Children, Toronto, Canada
| | - John Horan
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| |
Collapse
|
19
|
Sadowska-Klasa A, Piekarska A, Prejzner W, Bieniaszewska M, Hellmann A. Colonization with multidrug-resistant bacteria increases the risk of complications and a fatal outcome after allogeneic hematopoietic cell transplantation. Ann Hematol 2017; 97:509-517. [PMID: 29255911 PMCID: PMC5797223 DOI: 10.1007/s00277-017-3205-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
Composition of the gut microbiota seems to influence early complications of allogeneic hematopoietic cell transplantation (HCT) such as bacterial infections and acute graft-versus-host disease (GVHD). In this study, we assessed the impact of colonization with multidrug-resistant bacteria (MDRB) prior to HCT and the use of antibiotics against anaerobic bacteria on the outcomes of HCT. We retrospectively analyzed the data of 120 patients who underwent HCT for hematologic disorders between 2012 and 2014. Fifty-one (42.5%) patients were colonized with MDRB and 39 (32.5%) had infections caused by MDRB. Prior colonization was significantly correlated with MDRB infections (P < 0.001), especially bacteremia (P = 0.038). A higher incidence of MDRB infections was observed in patients with acute (P = 0.014) or chronic (P = 0.002) GVHD and in patients aged > 40 years (P = 0.002). Colonization had a negative impact on overall survival (OS) after HCT (64 vs. 47% at 24 months; P = 0.034) and infection-associated mortality (P < 0.001). Use of metronidazole was correlated with an increased incidence of acute GVHD (P < 0.001) and lower OS (P = 0.002). Patients colonized with MDRB are more susceptible to life-threatening infections. Colonization with virulent flora is the most probable source of neutropenic infection; therefore, information about prior positive colonization should be crucial for the selection of empiric antibiotic therapy. The use of metronidazole, affecting the biodiversity of the intestinal microbiome, seems to have a significant impact on OS and acute GVHD.
Collapse
Affiliation(s)
- Alicja Sadowska-Klasa
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| | - Agnieszka Piekarska
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland.
| | - Witold Prejzner
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| | - Maria Bieniaszewska
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| | - Andrzej Hellmann
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| |
Collapse
|
20
|
Group 3 innate lymphoid cells in tissue damage and graft-versus-host disease pathogenesis. Curr Opin Hematol 2017; 23:410-5. [PMID: 27135976 DOI: 10.1097/moh.0000000000000262] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW Innate lymphoid cells (ILC) have emerged as modulators of conditioning-induced tissue damage and development of graft-versus-host disease (GVHD) in the context of allogeneic hematopoietic stem cell transplantation (HSCT). This review highlights experimental and clinical evidence for a role of ILC in GVHD pathogenesis. RECENT FINDINGS ILC are well known for their role in epithelial homeostasis and innate immunity. In addition, recent studies identified ILC as architects of intestinal responses to tissue damage after experimental radio and chemotherapy. Group 3 ILC, and their signature cytokine IL-22, can enhance intestinal stem cell regeneration and protect the stem cell niche from damage during experimental HSCT. Moreover, in leukemia patients undergoing HSCT conditioning, appearance of activated group 3 ILC prior to transplant is correlated to reduced incidence of acute GVHD. SUMMARY ILC have a profound impact on the recovery from tissue damage and severity of GVHD in experimental models. Together with the available data from leukemia patients, this argues for in-depth analysis of the mechanisms of ILC function and the translation of experimental findings to clinical application. Ultimately, control of ILC activation, or of the cytokines they produce, could be employed to reduce GVHD lesion in patients receiving allogeneic HSCT.
Collapse
|
21
|
Dorshow RB, Hall-Moore C, Shaikh N, Talcott MR, Faubion WA, Rogers TE, Shieh JJ, Debreczeny MP, Johnson JR, Dyer RB, Singh RJ, Tarr PI. Measurement of gut permeability using fluorescent tracer agent technology. Sci Rep 2017; 7:10888. [PMID: 28883476 PMCID: PMC5589723 DOI: 10.1038/s41598-017-09971-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/31/2017] [Indexed: 12/27/2022] Open
Abstract
The healthy gut restricts macromolecular and bacterial movement across tight junctions, while increased intestinal permeability accompanies many intestinal disorders. Dual sugar absorption tests, which measure intestinal permeability in humans, present challenges. Therefore, we asked if enterally administered fluorescent tracers could ascertain mucosal integrity, because transcutaneous measurement of differentially absorbed molecules could enable specimen-free evaluation of permeability. We induced small bowel injury in rats using high- (15 mg/kg), intermediate- (10 mg/kg), and low- (5 mg/kg) dose indomethacin. Then, we compared urinary ratios of enterally administered fluorescent tracers MB-402 and MB-301 to urinary ratios of sugar tracers lactulose and rhamnose. We also tested the ability of transcutaneous sensors to measure the ratios of absorbed fluorophores. Urinary fluorophore and sugar ratios reflect gut injury in an indomethacin dose dependent manner. The fluorophores generated smooth curvilinear ratio trajectories with wide dynamic ranges. The more chaotic sugar ratios had narrower dynamic ranges. Fluorophore ratios measured through the skin distinguished indomethacin-challenged from same day control rats. Enterally administered fluorophores can identify intestinal injury in a rat model. Fluorophore ratios are measureable through the skin, obviating drawbacks of dual sugar absorption tests. Pending validation, this technology should be considered for human use.
Collapse
Affiliation(s)
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Michael R Talcott
- Division of Comparative Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - William A Faubion
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Roy B Dyer
- Immunochemical Core Laboratory, Mayo Clinic, Rochester, MN, USA
| | | | - Phillip I Tarr
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
22
|
The Impact of Toxicities on First-Year Outcomes after Ex Vivo CD34 +-Selected Allogeneic Hematopoietic Cell Transplantation in Adults with Hematologic Malignancies. Biol Blood Marrow Transplant 2017; 23:2004-2011. [PMID: 28733264 DOI: 10.1016/j.bbmt.2017.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/10/2017] [Indexed: 11/21/2022]
Abstract
Factors that impact first-year morbidity and mortality in adults undergoing myeloablative allogeneic hematopoietic cell transplantation with ex vivo CD34+ selection have not been previously reported. We assessed all toxicities ≥ grade 3 from the start of conditioning to date of death, relapse, or last contact in 200 patients during the first year after transplantation, identifying 1885 individual toxicities among 17 organ-based toxicity groups. The most prevalent toxicities in the first year were of infectious, metabolic, hematologic, oral/gastrointestinal, hepatic, cardiac, and pulmonary etiologies. Renal complications were minimal. Grades II to IV and III and IV acute GVHD at day 100 were 11.5% and 3%, respectively. In separate multivariate models, cardiovascular, hematologic, hepatic, neurologic, pulmonary, and renal toxicities negatively impacted nonrelapse mortality (NRM) and overall survival during the first year. A higher-than-targeted busulfan level, patient cytomegalovirus seropositivity, and an Hematopoietic Cell Transplantation-Specific Comorbidity Index of ≥3 were associated with increased risk of NRM and all-cause death. Ex vivo CD34+ selection had a favorable 1-year OS of 75% and NRM of 17% and a low incidence of sinusoidal obstruction syndrome. These data establish a benchmark to focus efforts in reducing toxicity burden while improving patient outcomes.
Collapse
|
23
|
Ranganathan P, Ngankeu A, Zitzer NC, Leoncini P, Yu X, Casadei L, Challagundla K, Reichenbach DK, Garman S, Ruppert AS, Volinia S, Hofstetter J, Efebera YA, Devine SM, Blazar BR, Fabbri M, Garzon R. Serum miR-29a Is Upregulated in Acute Graft-versus-Host Disease and Activates Dendritic Cells through TLR Binding. THE JOURNAL OF IMMUNOLOGY 2017; 198:2500-2512. [PMID: 28159900 DOI: 10.4049/jimmunol.1601778] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022]
Abstract
Acute graft-versus-host disease (aGVHD) continues to be a frequent and devastating complication of allogeneic hematopoietic stem cell transplantation (HSCT), posing as a significant barrier against the widespread use of HSCTs as a curative modality. Recent studies suggested serum/plasma microRNAs (miRs) may predict aGVHD onset. However, little is known about the functional role of circulating miRs in aGVHD. In this article, we show in two independent cohorts that miR-29a expression is significantly upregulated in the serum of allogeneic HSCT patients at aGVHD onset compared with non-aGVHD patients. Serum miR-29a is also elevated as early as 2 wk before time of diagnosis of aGVHD compared with time-matched control subjects. We demonstrate novel functional significance of serum miR-29a by showing that miR-29a binds and activates dendritic cells via TLR7 and TLR8, resulting in the activation of the NF-κB pathway and secretion of proinflammatory cytokines TNF-α and IL-6. Treatment with locked nucleic acid anti-miR-29a significantly improved survival in a mouse model of aGVHD while retaining graft-versus-leukemia effects, unveiling a novel therapeutic target in aGVHD treatment or prevention.
Collapse
Affiliation(s)
- Parvathi Ranganathan
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Apollinaire Ngankeu
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Nina C Zitzer
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - PierPaolo Leoncini
- Department of Oncohematology, Bambino Gesù Children's Hospital, Rome 00165, Italy
| | - Xueyan Yu
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Lucia Casadei
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Kishore Challagundla
- Department of Pediatrics, University of Southern California-Keck School of Medicine, Norris Comprehensive Cancer Center, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027
| | - Dawn K Reichenbach
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and
| | - Sabrina Garman
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Amy S Ruppert
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Stefano Volinia
- Department of Anatomy, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Jessica Hofstetter
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Yvonne A Efebera
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Steven M Devine
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and
| | - Muller Fabbri
- Department of Pediatrics, University of Southern California-Keck School of Medicine, Norris Comprehensive Cancer Center, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027
| | - Ramiro Garzon
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210;
| |
Collapse
|
24
|
Abstract
A fundamental function of the intestinal epithelium is to act as a barrier that limits interactions between luminal contents such as the intestinal microbiota, the underlying immune system and the remainder of the body, while supporting vectorial transport of nutrients, water and waste products. Epithelial barrier function requires a contiguous layer of cells as well as the junctions that seal the paracellular space between epithelial cells. Compromised intestinal barrier function has been associated with a number of disease states, both intestinal and systemic. Unfortunately, most current clinical data are correlative, making it difficult to separate cause from effect in interpreting the importance of barrier loss. Some data from experimental animal models suggest that compromised epithelial integrity might have a pathogenic role in specific gastrointestinal diseases, but no FDA-approved agents that target the epithelial barrier are presently available. To develop such therapies, a deeper understanding of both disease pathogenesis and mechanisms of barrier regulation must be reached. Here, we review and discuss mechanisms of intestinal barrier loss and the role of intestinal epithelial barrier function in pathogenesis of both intestinal and systemic diseases. We conclude with a discussion of potential strategies to restore the epithelial barrier.
Collapse
Affiliation(s)
- Matthew A Odenwald
- Department of Pathology, The University of Chicago, 5841 South Maryland, Chicago, Illinois 60637, USA
| | - Jerrold R Turner
- Department of Pathology, The University of Chicago, 5841 South Maryland, Chicago, Illinois 60637, USA
- Departments of Pathology and Medicine (Gastroenterology), Brigham and Women's Hospital and Harvard Medical School, 20 Shattuck Street, Thorn 1428, Boston, Massachusetts 02115, USA
| |
Collapse
|
25
|
Tu S, Zhong D, Xie W, Huang W, Jiang Y, Li Y. Role of Toll-Like Receptor Signaling in the Pathogenesis of Graft-versus-Host Diseases. Int J Mol Sci 2016; 17:E1288. [PMID: 27529218 PMCID: PMC5000685 DOI: 10.3390/ijms17081288] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/12/2016] [Accepted: 08/03/2016] [Indexed: 12/20/2022] Open
Abstract
Graft-versus-host disease (GVHD) and infection are major complications after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and the leading causes of morbidity and mortality in HSCT patients. Recent work has demonstrated that the two complications are interdependent. GVHD occurs when allo-reactive donor T lymphocytes are activated by major histocompatibility antigens or minor histocompatibility antigens on host antigen-presenting cells (APCs), with the eventual attack of recipient tissues or organs. Activation of APCs is important for the priming of GVHD and is mediated by innate immune signaling pathways. Current evidence indicates that intestinal microbes and innate pattern-recognition receptors (PRRs) on host APCs, including both Toll-like receptors (TLRs) and nucleotide oligomerization domain (NOD)-like receptors (NLRs), are involved in the pathogenesis of GVHD. Patients undergoing chemotherapy and/or total body irradiation before allo-HSCT are susceptible to aggravated gastrointestinal epithelial cell damage and the subsequent translocation of bacterial components, followed by the release of endogenous dangerous molecules, termed pathogen-associated molecular patterns (PAMPs), which then activate the PRRs on host APCs to trigger local or systemic inflammatory responses that modulate T cell allo-reactivity against host tissues, which is equivalent to GVHD. In other words, infection can, to some extent, accelerate the progression of GVHD. Therefore, the intestinal flora's PAMPs can interact with TLRs to activate and mature APCs, subsequently activate donor T cells with the release of pro-inflammatory cytokines, and eventually, induce GVHD. In the present article, we summarize the current perspectives on the understanding of different TLR signaling pathways and their involvement in the occurrence of GVHD.
Collapse
Affiliation(s)
- Sanfang Tu
- Department of Haematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Danli Zhong
- Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Weixin Xie
- Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Wenfa Huang
- Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Yangyang Jiang
- Department of Haematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Yuhua Li
- Department of Haematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
26
|
Rashidi A, Lin MF, Cashen AF. Early post-transplant contrast-enhanced abdominopelvic CT scan predicts the risk of subsequent acute GvHD. Bone Marrow Transplant 2015; 51:150-2. [PMID: 26437071 DOI: 10.1038/bmt.2015.232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- A Rashidi
- Section of Bone Marrow Transplant and Leukemia, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - M F Lin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - A F Cashen
- Section of Bone Marrow Transplant and Leukemia, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
27
|
Pontoppidan PEL, Shen RL, Cilieborg MS, Jiang P, Kissow H, Petersen BL, Thymann T, Heilmann C, Müller K, Sangild PT. Bovine Colostrum Modulates Myeloablative Chemotherapy-Induced Gut Toxicity in Piglets. J Nutr 2015; 145:1472-80. [PMID: 26019247 DOI: 10.3945/jn.114.203430] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/04/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Intensive chemotherapy frequently results in gut toxicity, indicated by oral and intestinal mucositis, resulting in poor treatment outcomes and increased mortality. There are no effective preventive strategies against gut toxicity and the role of diet is unknown. OBJECTIVE We hypothesized that the severity of chemotherapy-induced gut toxicity in early life is diet-dependent, and that intake of bovine colostrum (BC) provides better gut protection than an artificial milk replacer (MR). METHODS A total of 37 3-d-old pigs received for 6 d either intravenous saline control or myeloablative treatment with busulfan and cyclophosphamide, and were fed either BC or MR, resulting in the following 4 treatments (n = 8-10/group): bovine colostrum plus saline control (Ctr-BC), milk replacer plus saline control (Ctr-MR), bovine colostrum plus busulfan and cyclophosphamide chemotherapy (BUCY-BC), and milk replacer plus busulfan and cyclophosphamide chemotherapy (BUCY-MR). The gut was collected for analysis 11 d after the start of chemotherapy. RESULTS Relative to the control groups, both busulfan and cyclophosphamide chemotherapy (BUCY) groups showed signs of gut toxicity, with oral ulcers, reduced intestinal dimensions, and hematologic toxicity. Diet type did not affect mucosal structure on day 11, but BUCY-BC pigs had less vomiting than BUCY-MR pigs (1 of 10 vs. 10 of 10, P < 0.05). Markers of intestinal function were higher (up to 20-fold greater galactose absorption and 2-3-fold greater brush border enzyme activity, all P < 0.05), and tissue inflammatory cytokine concentrations and serum liver enzyme values were lower in BUCY-BC than in BUCY-MR pigs (30-50% reductions in interleukin 6 and 8, aminotransferase, and bilirubin concentrations, P < 0.05). Gut colonization was not significantly affected except that BUCY pigs had lower microbial diversity with a higher abundance of Lactobacilli. CONCLUSION BC may reduce gut toxicity during myeloablative chemotherapy in piglets by preserving intestinal function and reducing inflammation. Whether similar effects occur in children remains to be tested.
Collapse
Affiliation(s)
- Peter E L Pontoppidan
- Comparative Pediatrics and Nutrition, Department of Clinical Veterinary and Animal Science, University of Copenhagen, Frederiksberg, Denmark
| | - René L Shen
- Comparative Pediatrics and Nutrition, Department of Clinical Veterinary and Animal Science, University of Copenhagen, Frederiksberg, Denmark
| | - Malene S Cilieborg
- Comparative Pediatrics and Nutrition, Department of Clinical Veterinary and Animal Science, University of Copenhagen, Frederiksberg, Denmark
| | - Pingping Jiang
- Comparative Pediatrics and Nutrition, Department of Clinical Veterinary and Animal Science, University of Copenhagen, Frederiksberg, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Clinical Veterinary and Animal Science, University of Copenhagen, Frederiksberg, Denmark
| | | | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine and Institute of Inflammation Research, Department of Rheumatology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Per T Sangild
- Comparative Pediatrics and Nutrition, Department of Clinical Veterinary and Animal Science, University of Copenhagen, Frederiksberg, Denmark; Department of Pediatrics and Adolescent Medicine and
| |
Collapse
|
28
|
Intestinal barrier loss as a critical pathogenic link between inflammatory bowel disease and graft-versus-host disease. Mucosal Immunol 2015; 8:720-30. [PMID: 25943273 DOI: 10.1038/mi.2015.40] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/08/2015] [Indexed: 02/04/2023]
Abstract
Compromised intestinal barrier function is a prominent feature of inflammatory bowel disease (IBD). However, links between intestinal barrier loss and disease extend much further, including documented associations with celiac disease, type I diabetes, rheumatoid arthritis, and multiple sclerosis. Intestinal barrier loss has also been proposed to have a critical role in the pathogenesis of graft-versus-host disease (GVHD), a serious, potentially fatal consequence of hematopoietic stem cell transplantation. Experimental evidence has begun to support this view, as barrier loss and its role in initiating and establishing a pathogenic inflammatory cycle in GVHD is emerging. Here we discuss similarities between IBD and GVHD, mechanisms of intestinal barrier loss in these diseases, and the crosstalk between barrier loss and the immune system, with a special focus on natural killer (NK) cells. Unanswered questions and future research directions on the topic are discussed along with implications for treatment.
Collapse
|
29
|
Kuba A, Raida L, Mrazek F, Schneiderova P, Kriegova E, Furst T, Furstova J, Faber E, Ambruzova Z, Papajik T. ATM gene single nucleotide polymorphisms predict regimen-related gastrointestinal toxicity in patients allografted after reduced conditioning. Biol Blood Marrow Transplant 2015; 21:1136-40. [PMID: 25759145 DOI: 10.1016/j.bbmt.2015.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/23/2015] [Indexed: 10/23/2022]
Abstract
Polymorphisms of genes involved in innate and adaptive immunity have become an object of major interest in regard to hematopoietic stem cell transplantation (HSCT) complications. Regimen-related gastrointestinal toxicity (RR-GIT) is the dominant complication during the pre-engraftment period and has been linked to increased risk of graft-versus-host disease (GVHD) development. According to our hypothesis, functional variants of genes participating in DNA damage response (DDR) may have an impact on the extent of tissue damage caused by the conditioning regimen. In our single-center study, we analyzed 62 patients who underwent HSCT from HLA-identical donors after reduced conditioning. The patients were genotyped for 5 single nucleotide polymorphisms (SNPs, rs4585 T/G, rs189037 A/G, rs227092 T/G, rs228590 C/T, and rs664677 T/C) of the ATM gene-the essential member of the DDR pathways, using allele-specific matrix-assisted laser desorption/ionization, time-of-flight (MALDI-TOF) mass spectrometry assay. Because of almost absolute linkage disequilibrium observed among all 5 SNPs, association of 2 major ATM haplotypes (ATM1/ATM2) with RR-GIT and acute GVHD (aGVHD) was analyzed. Importantly, the univariate and multivariate analysis showed that patients homozygous for ATM2 haplotype (rs4585*T, rs189037*A, rs227092*T, rs228590*C, and rs664677*T) are more likely to suffer from high-grade RR-GIT than ATM1 homozygous patients. The association with aGVHD was not significant. To our knowledge, this is the first report showing the ATM gene variability in relation to RR-GIT in the allogeneic HSCT setting.
Collapse
Affiliation(s)
- Adam Kuba
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic.
| | - Ludek Raida
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Frantisek Mrazek
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petra Schneiderova
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Tomas Furst
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Jana Furstova
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Edgar Faber
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Zuzana Ambruzova
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Tomas Papajik
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
30
|
Nalle SC, Kwak HA, Edelblum KL, Joseph NE, Singh G, Khramtsova GF, Mortenson ED, Savage PA, Turner JR. Recipient NK cell inactivation and intestinal barrier loss are required for MHC-matched graft-versus-host disease. Sci Transl Med 2015; 6:243ra87. [PMID: 24990882 DOI: 10.1126/scitranslmed.3008941] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have shown a correlation between pretransplant conditioning intensity, intestinal barrier loss, and graft-versus-host disease (GVHD) severity. However, because irradiation and other forms of pretransplant conditioning have pleiotropic effects, the precise role of intestinal barrier loss in GVHD pathogenesis remains unclear. We developed GVHD models that allowed us to isolate the specific contributions of distinct pretransplant variables. Intestinal damage was required for the induction of minor mismatch [major histocompatibility complex (MHC)-matched] GVHD, but was not necessary for major mismatch GVHD, demonstrating fundamental pathogenic distinctions between these forms of disease. Moreover, recipient natural killer (NK) cells prevented minor mismatch GVHD by limiting expansion and target organ infiltration of alloreactive T cells via a perforin-dependent mechanism, revealing an immunoregulatory function of MHC-matched recipient NK cells in GVHD. Minor mismatch GVHD required MyD88-mediated Toll-like receptor 4 (TLR4) signaling on donor cells, and intestinal damage could be bypassed by parenteral lipopolysaccharide (LPS) administration, indicating a critical role for the influx of bacterial components triggered by intestinal barrier loss. In all, the data demonstrate that pretransplant conditioning plays a dual role in promoting minor mismatch GVHD by both depleting recipient NK cells and inducing intestinal barrier loss.
Collapse
Affiliation(s)
- Sam C Nalle
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - H Aimee Kwak
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Karen L Edelblum
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Nora E Joseph
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Gurminder Singh
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | | - Eric D Mortenson
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Peter A Savage
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| | - Jerrold R Turner
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
31
|
Costa-Lima C, De Paula EV. 'Leaky gut' in hematological malignancies. Rev Bras Hematol Hemoter 2014; 36:390-1. [PMID: 25453646 PMCID: PMC4318474 DOI: 10.1016/j.bjhh.2014.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 08/16/2014] [Indexed: 12/14/2022] Open
|
32
|
Vossen JM, Guiot HFL, Lankester AC, Vossen ACTM, Bredius RGM, Wolterbeek R, Bakker HDJ, Heidt PJ. Complete suppression of the gut microbiome prevents acute graft-versus-host disease following allogeneic bone marrow transplantation. PLoS One 2014; 9:e105706. [PMID: 25180821 PMCID: PMC4152127 DOI: 10.1371/journal.pone.0105706] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/22/2014] [Indexed: 11/18/2022] Open
Abstract
The hypothesis that elimination of facultative and strict anaerobic microorganisms from the gastro-intestinal tract by antimicrobial drugs in the period of time around allogeneic bone marrow transplantation (BMT) prevents acute graft-versus-host disease (GVHD), was examined in a cohort of 112 children grafted between 1989 and 2002 for hematological malignancies. All patients received T-cell replete marrow from human leukocyte antigens (HLA) matched sibling donors under identical transplantation conditions. To eliminate microorganisms from the gastro-intestinal tract, total gastro-intestinal decontamination (GID) was applied by high doses of non-absorbable antimicrobial drugs while the graft recipient was maintained in strict protective isolation. About half of the children (51%) proved to be successfully decontaminated, and about half (49%) unsuccessfully. One recipient got acute GVHD in the first group and 8 in the second group (p = 0.013). The degree of success of total GID was decisive for the occurrence of acute GVHD, irrespective of the presence of other risk factors such as higher age of recipient and/or donor, female donor for male recipient and carriership or reactivation of herpesviruses. Our results demonstrate that successful total GID of the graft recipient prevents moderate to severe acute GVHD. We suppose that substantial translocation of gastro-intestinal microorganisms or parts of these, functioning as microbial-associated molecular patterns (MAMP's), triggering macrophages/dendritic cells via pattern recognizing receptors (PRR's) is prohibited. As a consequence the initiation and progression of an inflammatory process leading to acute GVHD is inhibited.
Collapse
Affiliation(s)
- Jaak M. Vossen
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| | - Harry F. L. Guiot
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjan C. Lankester
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ann C. T. M. Vossen
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ron Wolterbeek
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hanny D. J. Bakker
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter J. Heidt
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| |
Collapse
|
33
|
Souza NCS, Simões BP, Júnior AAJ, Chiarello PG. Changes in Intestinal Permeability and Nutritional Status After Cytotoxic Therapy in Patients with Cancer. Nutr Cancer 2014; 66:576-82. [DOI: 10.1080/01635581.2014.894095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Abstract
β-TrCP, the substrate recognition subunit of SCF-type ubiquitin ligases, is ubiquitously expressed from two distinct paralogs, targeting for degradation many regulatory proteins, among which is the NF-κB inhibitor IκB. To appreciate tissue-specific roles of β-TrCP, we studied the consequences of inducible ablation of three or all four alleles of the E3 in the mouse gut. The ablation resulted in mucositis, a destructive gut mucosal inflammation, which is a common complication of different cancer therapies and represents a major obstacle to successful chemoradiation therapy. We identified epithelial-derived IL-1β as the culprit of mucositis onset, inducing mucosal barrier breach. Surprisingly, epithelial IL-1β is induced by DNA damage via an NF-κB-independent mechanism. Tissue damage caused by gut barrier disruption is exacerbated in the absence of NF-κB, with failure to express the endogenous IL-1β receptor antagonist IL-1Ra upon four-allele loss. Antibody neutralization of IL-1β prevents epithelial tight junction dysfunction and alleviates mucositis in β-TrCP-deficient mice. IL-1β antagonists should thus be considered for prevention and treatment of severe morbidity associated with mucositis.
Collapse
|
35
|
Merlin E, Minet-Quinard R, Pereira B, Rochette E, Auvrignon A, Oudot C, Sapin V, Deméocq F, Kanold J. Non-invasive biological quantification of acute gastrointestinal graft-versus-host disease in children by plasma citrulline. Pediatr Transplant 2013; 17:683-7. [PMID: 23901800 DOI: 10.1111/petr.12128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/27/2013] [Indexed: 01/22/2023]
Abstract
Clinical grading of GI involvement during acute GVHD remains a challenging issue, especially in children. Plasma citrulline, a non-protein amino acid selectively produced and released by enterocytes, is a suitable surrogate endpoint for small intestinal epithelial cell mass, irrespective of the underlying cause of cell loss. Children referred for allogeneic bone marrow transplantation who were free from chronic malabsorption or constitutional disease involving the GI tract were consecutively included in this prospective study. Plasma citrulline and albumin concentration was measured every week between day 7 and day 28 of BMT until resolution of the aGVHD or occurrence of chronic GVHD. In total, 31 children were included between 2008 and 2011. After a CR, citrulline levels fell to a minimum level on day 7 and then increased to reach the initial value on day 28. After day 28, plasma citrulline but not albumin was strongly linked to the occurrence of GI GVHD, the threshold being set at 10 μmol/L. The correlation with clinical grade of GI-aGVHD now needs to be assessed in larger populations. In pediatric patients, citrulline is valuable as a suitable non-invasive marker of GI involvement in acute GVHD.
Collapse
Affiliation(s)
- E Merlin
- Centre Régional de Cancérologie et Thérapie Cellulaire Pédiatrique, CHU Clermont-Ferrand, CHU Estaing, Clermont-Ferrand, France; INSERM, CIC 501, Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
An essential role of the intestinal epithelium is to separate luminal contents from the interstitium, a function primarily determined by the integrity of the epithelium and the tight junction that seals the paracellular space. Intestinal tight junctions are selectively permeable, and intestinal permeability can be increased physiologically in response to luminal nutrients or pathologically by mucosal immune cells and cytokines, the enteric nervous system, and pathogens. Compromised intestinal barrier function is associated with an array of clinical conditions, both intestinal and systemic. Although most available data are correlative, some studies support a model where cycles of increased intestinal permeability, intestinal immune activation, and subsequent immune-mediated barrier loss contribute to disease progression. This model is applicable to intestinal and systemic diseases. However, it has not been proven, and both mechanistic and therapeutic studies are ongoing. Nevertheless, the correlation between increased intestinal permeability and disease has caught the attention of the public, leading to a rise in popularity of the diagnosis of "leaky gut syndrome," which encompasses a range of systemic disorders. Proponents claim that barrier restoration will cure underlying disease, but this has not been demonstrated in clinical trials. Moreover, human and mouse studies show that intestinal barrier loss alone is insufficient to initiate disease. It is therefore uncertain whether increased permeability in these patients is a cause or effect of the underlying disorder. Although drug targets that may mediate barrier restoration have been proposed, none have been proven effective. As such, current treatments for barrier dysfunction should target the underlying disease.
Collapse
Affiliation(s)
- Matthew A Odenwald
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
37
|
Blennow O, Mattsson J, Remberger M. Pre-engraftment blood stream infection is a risk factor for acute GVHD grades II–IV. Bone Marrow Transplant 2013; 48:1583-4. [DOI: 10.1038/bmt.2013.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
Mårtensson T, Priftakis P, Casswall T, Ringdén O, Mattsson J, Remberger M, Hassan M, Gustafsson B. Increased risk of gastrointestinal acute GVHD following the addition of melphalan to busulfan/cyclophosphamide conditioning. Pediatr Transplant 2013; 17:285-93. [PMID: 23489519 DOI: 10.1111/petr.12061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2013] [Indexed: 11/30/2022]
Abstract
Risk factors associated with the development of aGVHD in the gastrointestinal tract have not been studied in depth. We retrospectively assessed 25 pediatric patients with MDS and JMML and compared the treatment outcome of two different conditioning regimens. Seventeen children (68%) underwent conditioning with busulfan (Bu), cyclophosphamide (Cy), and melphalan (Mel) and eight children (32%) with Bu and Cy. Gastrointestinal aGVHD stages II-IV (day 0-100) were observed in 47% (eight of 17) of the patients in the BuCyMel group and in none (0 of 8) in the BuCy group (p < 0.05). In patients who developed gastrointestinal aGVHD stages III-IV, a 24-h variation in the Bu concentration with a nighttime peak was noted. HC and liver aGVHD stages II-IV were observed in 47% (eight of 17) and 35% (six of 17) after BuCyMel conditioning and in 0% (0 of 17) and 12.5% (one of eight) after BuCy conditioning. The overall survival rate was 53% (nine of 17) in the BuCyMel group and 62.5% (five of eight) in the BuCy group. In conclusion, the addition of melphalan to the BuCy conditioning regimen resulted in severe gastrointestinal complications and did not improve overall survival.
Collapse
Affiliation(s)
- T Mårtensson
- Astrid Lindgren Children's Hospital and Karolinska University Hospital-Huddinge, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Endothelial and epithelial barriers in graft-versus-host disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 763:105-31. [PMID: 23397621 DOI: 10.1007/978-1-4614-4711-5_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Endothelial and epithelial cells form selectively permeable barriers that separate tissue compartments. These cells coordinate movement between the lumen and tissue via the transcellular and paracellular pathways. The primary determinant of paracellular permeability is the tight junction, which forms an apical belt-like structure around endothelial and epithelial cells. This chapter discusses endothelial and epithelial barriers in graft-versus-host disease after allogeneic bone marrow transplantation, with a focus on the tight junction and its role in regulating paracellular permeability. Recent studies suggest that in graft-versus-host disease, pathological increases in paracellular permeability, or barrier dysfunction, are initiated by pretransplant conditioning and sustained by alloreactive cells and the proinflammatory milieu. The intestinal epithelium is a significant focus, as it is a target organ of graft-versus-host disease, and the mechanisms of barrier regulation in intestinal epithelium have been well characterized. Finally, we propose a model that incorporates endothelial and epithelial barrier dysfunction in graft-versus-host disease and discuss modulating barrier properties as a therapeutic approach.
Collapse
|
40
|
Poutsiaka DD, Munson D, Price LL, Chan GW, Snydman DR. Blood stream infection (BSI) and acute GVHD after hematopoietic SCT (HSCT) are associated. Bone Marrow Transplant 2011; 46:300-7. [PMID: 20479711 PMCID: PMC3049187 DOI: 10.1038/bmt.2010.112] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 03/15/2010] [Accepted: 03/17/2010] [Indexed: 01/27/2023]
Abstract
Blood stream infection (BSI) and acute GVHD (aGVHD) are serious complications of hematopoietic SCT (HSCT). We hypothesized that the two events were not independent of one another. We studied (1) associations between BSI and aGVHD; and (2) the impact of BSI and/or aGVHD on death within 100 days after HSCT, using a retrospective cohort analysis. Risk factor analysis was carried out using multivariable Cox proportional hazards analyses. Of 211 patients who underwent allogeneic HSCT from January 2000 to December 2005 (58% of whom underwent reduced intensity transplantation), 82 (39%) developed BSI. In 49 patients (23%), grade (gr) 2-4 aGVHD occurred. Early BSI was independently associated with an increased occurrence of subsequent aGVHD gr 2-4. CMV seropositivity was independently associated with decreased occurrence of aGVHD. aGVHD gr 2-4 independently predicted subsequent first BSI. Both BSI and aGVHD gr 2-4 were significant independent predictors of death within 100 days after HSCT. There is a strong, independent association between BSI and aGVHD. Potential explanations include the elaboration of cytokines during BSI favoring the development of aGVHD and/or the immunosuppressive treatment of aGVHD favoring the development of BSI. Future studies should be directed at the mechanistic investigations of this association.
Collapse
Affiliation(s)
- D D Poutsiaka
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA 02111, USA.
| | | | | | | | | |
Collapse
|
41
|
Holler E, Landfried K, Meier J, Hausmann M, Rogler G. The role of bacteria and pattern recognition receptors in GVHD. Int J Inflam 2010; 2010:814326. [PMID: 21188220 PMCID: PMC3003997 DOI: 10.4061/2010/814326] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 09/02/2010] [Indexed: 01/01/2023] Open
Abstract
Graft-versus-Host Disease (GvHD) is the most serious complication of allogeneic stem cell transplantation (SCT) and results from an activation of donor lymphocytes by recipient antigen-presenting cells (APCs). For a long time, it has been postulated that the intestinal microflora and endotoxin exert a crucial step in this APC activation, as there is early and severe gastrointestinal damage induced by pretransplant conditioning. With the detailed description of pathogen-associated molecular patterns and pathogen recognition receptors single nucleotide polymorphisms of TLRs and especially NOD2 have been identified as potential risk factors of GvHD and transplant related complications thus further supporting the crucial role of innate immunity in SCT, related complications. Gastrointestinal decontamination and neutralization of endotoxin have been used to interfere with this early axis of activation with some success but more specific approaches of modulation of innate immunity are needed for further improvement of clinical outcome.
Collapse
Affiliation(s)
- E Holler
- Department of Haematology/Oncology, University of Regensburg, 93042 Regensburg, Germany
| | | | | | | | | |
Collapse
|
42
|
Maingat F, Viappiani S, Zhu Y, Vivithanaporn P, Ellestad KK, Holden J, Silva C, Power C. Regulation of lentivirus neurovirulence by lipopolysaccharide conditioning: suppression of CXCL10 in the brain by IL-10. THE JOURNAL OF IMMUNOLOGY 2009; 184:1566-74. [PMID: 20042580 DOI: 10.4049/jimmunol.0902575] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lentivirus infections including HIV and feline immunodeficiency virus (FIV) cause neurovirulence, which is largely mediated by innate immunity. To investigate the interactions between neurovirulence and repeated conditioning by innate immune activation, models of lentivirus infection were exposed to LPS. Gene expression in HIV-infected (HIV+) and control (HIV-) patient brains was compared by real time RT-PCR and immunocytochemistry. Supernatants from mock and HIV-infected monocyte-derived macrophages exposed to LPS were applied to human neurons. FIV-infected (FIV+) and control (FIV-) animals were exposed repeatedly to LPS postinfection together with concurrent neurobehavioral testing, viral load, and host gene analyses. Brains from HIV+ individuals exhibited induction of CD3epsilon, CXCL10, and granzyme A expression (p < 0.05). Supernatants from HIV+ monocyte-derived macrophages induced CXCL10 expression in neurons, which was diminished by IL-10 treatment (p < 0.05). LPS-exposed FIV+ animals demonstrated lower plasma and brain viral loads (p < 0.05). Neuronal CXCL10 expression was increased in FIV+ animals but was suppressed by LPS exposure, together with reduced brain CD3epsilon and granzyme A expression (p < 0.05). In conjunction with preserved NeuN-positive neuronal counts in parietal cortex (p < 0.05), FIV+ animals exposed to LPS also showed less severe neurobehavioral deficits (p < 0.05). Repeated LPS exposures suppressed CXCL10 in the brain and ensuing T cell infiltration with a concomitant reduction in neurovirulence. Thus, innate immune chronic conditioning exerted beneficial effects on neurovirulence through suppression of a specific chemotactic factor, CXCL10, mediated by IL-10, leading to reduced leukocyte infiltration and release of neurotoxic factors.
Collapse
|
43
|
Johansson JE, Hasséus B, Johansson P, Eklöf C, Ohman D, Stockelberg D. Gut protection by palifermin during autologous haematopoietic SCT. Bone Marrow Transplant 2008; 43:807-11. [PMID: 19043460 DOI: 10.1038/bmt.2008.388] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Conditioning therapy in connection with haematopoietic SCT (HSCT) induces a disruption of the intestinal barrier function facilitating the permeation of bacteria and endotoxin through the bowel wall with subsequent increased risk of septicaemia and a worsening of GVHD in the allogeneic setting. Palifermin (recombinant human keratinocyte growth factor) reduces the severity of oral mucositis with HSCT. The present trial investigates its effect on intestinal barrier function. Seventeen lymphoma patients undergoing autologous HSCT received palifermin. Intestinal permeability was assessed before the conditioning therapy and on days +4 and +14. Clinical oral and gastrointestinal toxicity was prospectively assessed in parallel. A comparison was made with matched historical study patients (n=21). Patients treated with palifermin had a significantly better preserved intestinal barrier function (P=0.01 on day +4) and were in less need of total parenteral nutrition (P=0.005) as compared with controls. No significant reduction of clinical gastrointestinal or oral toxicity was observed. The intestinal barrier function, normally disrupted by the conditioning therapy, is preserved by palifermin. Whether intestinal barrier preservation protects from invasive infections, and in the allogeneic setting diminishes GVHD severity, remains to be investigated in randomized controlled trials.
Collapse
Affiliation(s)
- J-E Johansson
- Department of Haematology, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | | | | | | | | | |
Collapse
|