ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathogenesis of graft-versus-host disease (GVHD)

Pathogenesis of graft-versus-host disease (GVHD)
Literature review current through: Jan 2024.
This topic last updated: May 04, 2023.

INTRODUCTION — Graft-versus-host disease (GVHD) is the major cause of morbidity and non-relapse mortality in patients after allogeneic hematopoietic cell transplantation (HCT). GVHD refers to multi-organ syndromes of tissue inflammation and/or fibrosis that primarily affect skin, gastrointestinal tract, liver, lungs, and mucosal surfaces. Clinically, GVHD comprises three syndromes, acute GVHD (aGVHD), chronic GVHD (cGVHD), and GVHD overlap syndrome. The various GVHD syndromes are defined by clinical manifestations according to National Institutes of Health consensus criteria [1], rather than the time of onset (ie, before or after day 100 of transplantation, as was used previously). Greater understanding of the underlying pathophysiology is important for more effectively controlling GVHD and improving clinical outcomes with HCT.

This topic will discuss the pathogenesis of aGVHD and cGVHD.

Clinical manifestations, diagnosis, grading, treatment, and prevention of aGVHD and cGVHD are discussed separately. (See "Clinical manifestations, diagnosis, and grading of acute graft-versus-host disease" and "Clinical manifestations and diagnosis of chronic graft-versus-host disease" and "Prevention of graft-versus-host disease" and "Treatment of chronic graft-versus-host disease".)

Innate immunity, transplantation immunobiology, and normal B and T lymphocyte development are discussed separately. (See "An overview of the innate immune system" and "Transplantation immunobiology" and "Normal B and T lymphocyte development".)

OVERVIEW OF GVHD — GVHD refers to multi-organ syndromes that can develop after allogeneic hematopoietic cell transplantation (HCT). GVHD arises from one of the principal functions of the immune system: distinguishing between self and non-self. GVHD occurs when immune cells transplanted from a non-identical donor (graft) into the recipient (host) recognize the host cells as "foreign," thereby initiating a graft-versus-host reaction [2,3]. Successful transplantation requires that the donor immune system develop tolerance to these alloantigens, while maintaining the ability to recognize and respond to foreign antigens, such as microorganisms or tumor cells.

GVHD is manifest clinically as three syndromes with different clinical manifestations and temporal courses:

Acute GVHD (aGVHD) – Characterized by a rapid onset and acute disease course

Chronic GVHD (cGVHD) – Characterized by a chronic disease course that can involve virtually all organs with variable manifestations, including sclerosis

GVHD overlap syndrome – Defined by simultaneous features of both cGVHD and aGVHD

Both aGVHD and cGVHD are consequences of the interplay between cellular/immune mediators from the immunological graft with host tissues. Although the two syndromes share some features, they differ with regard to aspects of the underlying pathophysiology, pathology, clinical manifestations, and management. Whereas aGVHD is typically manifest as an inflammatory immune cell infiltrate, including T cells, neutrophils, and monocytes, with tissue destruction, the tissue response in cGVHD is relatively acellular and reveals fibroproliferative findings. Acute GVHD is primarily driven by activation of donor T lymphocytes and release of pro-inflammatory cytokines; by contrast, cGVHD is a more complex and less well-understood syndrome that involves interactions of the innate immune system (macrophages, neutrophils, dendritic cells) with alloreactive and dysregulated B and T cells and non-hematopoietic cells, such as fibroblasts. Further details of the pathophysiologic mechanisms that underlie aGVHD and cGVHD are provided in the sections below. (See 'Acute GVHD' below and 'Chronic GVHD' below.)

It is unclear if the GVHD effect can be separated from the graft-versus-tumor effect. (See "Biology of the graft-versus-tumor effect following hematopoietic cell transplantation", section on 'Possible separation of GVT from GVHD'.)

PATHOPHYSIOLOGY — Greater understanding of the pathophysiology is important for development of new and more effective treatments for GVHD.

Acute GVHD — Acute GVHD (aGVHD) is primarily manifest as a maculopapular rash, weight loss, diarrhea, and/or hepatitis that typically occurs within the first 100 days after transplantation. Clinical manifestations of aGVHD are discussed separately. (See "Clinical manifestations, diagnosis, and grading of acute graft-versus-host disease", section on 'Clinical and histological manifestations'.)

Pathologically, aGVHD is apparent as an inflammatory immune cell infiltrate involving T cells, macrophages, monocytes and neutrophil granulocytes with associated tissue destruction and apoptosis. The transplantation conditioning regimen, innate immune system, and gastrointestinal (GI) microbiome all contribute to the pathophysiology of aGVHD [4,5]:

Conditioning regimen – The transplantation conditioning regimen damages the GI epithelium and leads to translocation of bacteria, fungi, and viruses that initiate inflammation mediated by the innate immune system in cooperation with T and B lymphocytes of the adaptive immune system [6,7]. Macrophages, neutrophils, and dendritic cells mediate this response through Toll-like receptors (TLR) and through nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs), which are involved in pathogen recognition via patterns of nucleic acids or sugars [8-11]. Non-pathogen associated molecules such as uric acid, adenosine triphosphate, or IL-33 (so-called damage associated molecular patterns [DAMPs]) also promote aGVHD [12,13]. Cellular components of the innate immune system and TLRs are discussed separately. (See "An overview of the innate immune system" and "Toll-like receptors: Roles in disease and therapy".)

Inflammatory milieu – A pro-inflammatory milieu activates antigen presenting cells (APC) that prime naïve T cells to Th1 and Th17 differentiation, expand T effector cells, and target host tissues. Scavenger macrophages, plasmacytoid and myeloid dendritic cells, B cells, and neutrophils produce cytokines that enhance antigen presentation and drive differentiation to the Th1 and Th17 effector lineages [6]. Signaling through Janus kinase (JAK)1 and JAK2 and signal transducers and activators of transcription (STAT) contributes to inflammation and tissue damage by neutrophils, dendritic cells, and inflammatory cytokines [14-18]. TLR pathway activation induces transcriptional activation of interferon (IFN) alpha (IFNa) through IFN response factors (IRF 3, IRF 7) and induces tumor necrosis factor (TNF) and interleukin (IL)-6 through nuclear factor kappa B (NFkB) [19-21]. IFNa can drive Th1 commitment and result in IFN gamma (IFNg) production and, together, IFNa and IFNg induce chemokines (eg, CXCL9, CXCL10, CXCL11) that recruit Th1 cells to sites of inflammation and enhance processing and presentation of host antigens [6,22]. Inflammasome complexes catalyze production of IL-1b and IL-18 which, together with IL-6, induce differentiation of Th17 cells, regulate antigen presentation and migration of dendritic cells and lymphocytes, and result in loss of myeloid-derived suppressor cell function [23,24]. Human beta-defensin 2 (hBD-2), an endogenous epithelial cell-derived host-defense peptide, was shown to reduce inflammation in mice developing aGVHD [25].

Tissue regeneration – Acute GVHD also affects enterocytes, intestinal stem cells (ISC), and Paneth cells and dysregulates tissue-regenerative mechanisms such as intestinal stem cell (ISC) and Paneth cell repair; these effects may enable therapeutic strategies to enhance intestinal cell repair (eg, treatment with IL-22, R-spondin, keratinocyte growth factor, and Glucagon-like peptide 2) [26-29].

Experimental models and clinical experience confirm the importance of innate immunity and TLRs in aGVHD. Deletion or inhibition of TLR or NOD-like receptor pathways significantly reduce aGVHD [30-33]. Furthermore, polymorphisms of proteins that mediate innate immunity are associated with clinical outcomes in HCT [34]. As an example, NOD2/CARD15 is an intracellular sensor of muramyl dipeptide (a component of the bacterial cell wall) that is expressed by intestinal epithelial cells and cells of monocyte/macrophage lineage and mediates activation of NFkB. In a study of 169 consecutive patients receiving transplants from related or unrelated donors, polymorphisms of NOD2/CARD15 were found in 21 percent of recipients and 14 percent of donors [34]. The cumulative incidence of one-year transplant-related mortality rose from 20 percent in donor/recipient pairs without single nucleotide polymorphisms, to 49 percent in pairs with recipient mutations, 59 percent in pairs with donor mutations, and 83 percent in 12 pairs with mutated alleles in both donor and recipient.

Observational studies suggest that the diversity and composition of the GI microbiome (ie, intestinal bacteria) plays a role in the development of GVHD involving the lower GI tract, as discussed in more detail separately.

Chronic GVHD — Chronic GVHD (cGVHD) is manifest as fibrosis and chronic inflammation of skin, lungs, GI tract, and soft tissues that generally presents ≥100 days after transplantation. Clinical aspects of cGVHD are discussed separately. (See "Clinical manifestations and diagnosis of chronic graft-versus-host disease", section on 'Clinical manifestations'.)

Pathologically, tissues affected by cGVHD are relatively acellular and fibroproliferative. Development of cGVHD is a complex, multi-phase process that involves various cell lineages and types of injury [6,19,35]. In cGVHD, early inflammation results from the conditioning regimen and activation of donor T cells. Injury of vascular endothelial cells (EC) facilitates migration of donor immune cells into target organs. Donor-derived effector T lymphocytes, B lymphocytes, and APCs mount an immune response against host tissues. Immune tolerance is affected by depletion of regulatory T cells (Treg) and their functional suppression by calcineurin inhibitors [36], along with thymic injury/dysfunction [37]. Aberrant repair mechanisms foster activation of fibroblasts, collagen deposition, and fibrosis that lead to irreversible end-organ injury and dysfunction.

Experimental studies support a three-phase model of cGVHD [6]:

Early inflammation and tissue injury – Early inflammation and tissue injury in cGVHD is initiated and sustained by the innate immune system. The cellular components (ie, macrophages, neutrophils, dendritic cells, and B cells), signaling mechanisms (eg, TLR and NOD-like pathways), and mediators (eg, cytokines) of cGVHD resemble the mechanisms that underlie aGVHD [6], as described above. (See 'Acute GVHD' above.)

Activation and injury of ECs contribute to early inflammation in cGVHD [6]. ECs function as a barrier between donor and recipient tissues and they are the first host cells encountered by the transplanted donor immune system. EC injury and early inflammation may be caused by irradiation, lipopolysaccharide, TNFa, and cytotoxic lymphocytes.

Mature donor T cells infused with the transplanted host graft also contribute to inflammation. Depletion of T cells in vivo or short courses of cyclophosphamide reduce the incidence and severity of cGVHD, which supports this observation [6]. Activation and clonal expansion of donor T cells into Th2 and Th17 functional subsets produces inflammatory cytokines and cytolytic enzymes that contribute to the early inflammation of cGVHD. Mobilization of peripheral blood stem cells with granulocyte colony-stimulating factor (G-CSF) also promotes Th17 differentiation [38].

Chronic inflammation and tissue injury – Donor- and/or host-derived immune regulatory responses are not sufficient to control the early inflammation, which results in chronic inflammation and dysregulated immunity [6]. Tregs are important for immune homeostasis and immune tolerance, and dysregulated Tregs contribute to sustained inflammation in cGVHD, although the underlying mechanisms are poorly defined [6,39,40]. Effects of early inflammation on the thymus may also contribute to a lack of immune tolerance and sustained inflammation in cGVHD [41]. Disordered immune suppression by type 1 regulatory T cells (Tr1), myeloid-derived suppressor cells, and other cell types also contribute to persistent inflammation [42-44].

Diminished immune regulatory functions of B cells and natural killer (NK) cells also contribute to chronic inflammation. Detection of autoantibodies against minor histocompatibility antigens, antinuclear antibodies (ANA), anti-double-stranded DNA indicate a loss of B cell tolerance [45]. NK cells are cytotoxic lymphocytes that express killer-cell immunoglobulin-like receptors (KIR), which can detect major histocompatibility complex (MHC) on the cell surface, trigger cytokine release, and cause lysis or apoptosis of target cells. KIR haplotypes can be activating or inhibitory. (See "An overview of the innate immune system", section on 'Natural killer cells'.)

Aberrant tissue repair and fibrosis – Dysregulated immunity and aberrant tissue repair contribute to scarring and fibrosis in cGVHD [6]. Early EC damage activates coagulation pathways that release chemotactic factors, and macrophages are a source of transforming growth factor (TGF) beta (TGFb), TNFa, IL-1b, platelet-derived growth factor (PDGF), and matric metalloproteinases, with an ensuing cascade of fibrosis [46]. IL-22 may also contribute to cutaneous manifestations of cGVHD [47]. Fibroblasts contribute to extracellular matrix production and collagen deposition.

Adaptive immunity also contributes to tissue injury and scarring. Activated Th2 and Th17 T cells promote fibrosis by secretion of IL-13 and IL-17, respectively [6]. B cell activation contributes auto- and allo-antibody production which, in concert with colony-stimulating factor 1 (CSF-1), further activate monocytes and macrophages to release TGFb, which activates myofibroblasts and collagen production leading to further tissue scarring and fibrosis [48].

CONTRIBUTING FACTORS

Histocompatibility — GVHD arises when immune cells transplanted from a non-identical graft recognize cells in the host as foreign. The major histocompatibility complex (MHC) provides the crucial surface upon which foreign antigens are displayed for immune recognition by T lymphocytes. Minor antigens also contribute to tissue histocompatibility. The MHC and mechanisms of allorecognition are discussed separately. (See "Transplantation immunobiology".)

MHC/HLA antigens — In humans, MHC molecules are called human leukocyte antigens (HLA). HLA is highly polymorphic from individual to individual and segregates in families in a Mendelian codominant fashion. (See "Transplantation immunobiology", section on 'Major histocompatibility complex structure and function'.)

In allogeneic hematopoietic cell transplantation (HCT), the principal antigenic targets of the T cells of the graft are host MHC molecules. The genes of the HLA locus encode two distinct classes of cell surface molecules, class I and class II. There are three different class I (HLA-A, -B, -C) and class II (HLA-DQ, -DR, -DP) antigens. HLA-A, -B and -DR antigens appear to be the most important loci for determining whether transplanted cells initiate a graft-versus-host reaction [49]. Class I molecules are expressed on the surfaces of virtually all nucleated cells at varying densities, while class II molecules are more restricted to cells of the immune system, primarily B lymphocytes and monocytes. However, cytokines secreted by lymphocytes and monocytes during immune activation may cause dramatic increases in class II HLA antigen expression, even on cell types that normally have little or no surface expression. (See "Human leukocyte antigens (HLA): A roadmap".)

Antigen-presenting cells, such as macrophages, present a complex of an MHC molecule bearing a small peptide fragment to a lymphocyte, which expresses a single T cell receptor (TCR). Class II molecules display antigenic peptide fragments to CD4-positive inducer (helper) T cells. Class I molecules function at the effector phase of immunity by presenting antigens to CD8-positive T cells, which generally have cytotoxic/suppressor function. (See "Transplantation immunobiology".)

The role of MHC/HLA in selection of a donor for HCT is discussed separately. (See "Donor selection for hematopoietic cell transplantation".)

Minor histocompatibility antigens — GVHD can develop even with grafts that are fully matched at the MHC/HLA loci due to mismatching of other antigens, termed minor histocompatibility antigens (miH).

Minor antigens (miH) are presented in the context of MHC. Because the manner in which a particular protein is processed is dependent upon genes outside of the MHC, two siblings, despite having identical MHC molecules, will have different peptides in the MHC groove [50,51]. MHC class I-related chain A (MICA) and killer-cell immunoglobulin-like receptor (KIR) are examples of miH that can cause rejection. Other specific proteins that account for miH in humans are poorly defined. (See "Transplantation immunobiology", section on 'Minor transplantation antigens'.)

Histocompatibility antigen matching — HLA can be matched serologically or using genetic-based testing, as discussed separately. (See "Human leukocyte antigens (HLA): A roadmap".)

Tissue microenvironment — Cells, cytokines, and signaling pathways of the tissue microenvironment contribute to GVHD and the graft-versus-leukemia (GVL) effect.

As an example, Notch signaling, which orchestrates cell fate and differentiation, is critical in both acute and chronic GVHD. Notch inhibition results in blockade of multiple cytokines, expansion of T regs and decrease in pathogenic T cells without decreasing GVL [52]. Similarly, T follicular helper cells in germinal centers of secondary lymphoid organs contribute to development of GVHD [53].

Clinical factors — A number of clinical variables are associated with the development of GVHD and may influence the underlying pathophysiology [6]. Factors that relate to clinical features of the recipient and the donor are discussed separately. (See "Clinical manifestations, diagnosis, and grading of acute graft-versus-host disease", section on 'Risk factors' and "Donor selection for hematopoietic cell transplantation".)

Clinical factors that contribute to GVHD include:

Donor type (ie, matched related, matched unrelated, haploidentical)

Source (peripheral blood, bone marrow, umbilical cord)

Sex-mismatch

Age of donor and recipient

Conditioning regimen intensity

Underlying malignancy (eg, myelodysplastic syndrome, acute or chronic myeloid or lymphoid leukemia)

T cell depletion in vivo (eg, anti-thymocyte globulins, alemtuzumab)

Post-transplantation cyclophosphamide

Infection history (eg, CMV, EBV)

The roles of these factors in selection of a HCT donor are discussed separately. (See "Donor selection for hematopoietic cell transplantation", section on 'Effect of donor characteristics'.)

Microbiome — The composition of gastrointestinal microbiota has been associated with outcomes in patients who undergo allogeneic HCT. However, it is not clear that this is a causal relationship or if it is possible to manipulate the intestinal microbiome to influence outcomes.

An increase of potentially pathogenic bacteria and loss of diversity in the number of bacterial taxa is commonly found in patients undergoing allogeneic HCT [54-57]. A large international study reported that higher diversity of intestinal microbiota was associated with lower mortality, lower rates of transplant-related death, and fewer deaths attributable to GVHD [58]. The study profiled 8767 fecal samples from 1362 patients at four institutions and used 16S ribosomal RNA sequence to stratify patients into higher-diversity (HD) and lower-diversity (LD) groups. In a preliminary study at one of the institutions, compared with LD patients, patients with HD had a hazard ratio (HR) for death of 0.71 (95% CI 0.55-0.92); analysis from three other institutions reported the HR for death was 0.49 (95% CI 0.27-0.90). Samples obtained before transplantation already showed evidence of microbiome disruption, and lower diversity before transplantation was also associated with poor survival. Single-institution studies have reported similar associations between diversity of intestinal microbiota and transplantation outcomes [59-61].

SUMMARY

Description – Graft-versus-host disease (GVHD) is the major source of non-relapse morbidity and mortality in patients who undergo allogeneic hematopoietic cell transplantation (HCT). GVHD refers to multi-organ syndromes of tissue inflammation and/or fibrosis that primarily affect skin, gastrointestinal tract, liver, lungs, and mucosal surfaces. Greater understanding of the pathophysiology of GVHD is important for development of new and more effective treatments.

Overview – GVHD arises when immune cells transplanted from a non-identical donor (graft) into the recipient (host) recognize the host cells as "foreign," thereby initiating a graft-versus-host reaction. GVHD is manifest clinically as three syndromes that are generally defined by their clinical manifestation (see 'Overview of GVHD' above):

GVHD comprises three syndromes:

Acute GVHD (aGVHD) – Characterized by a rapid onset and acute disease course

Chronic GVHD (cGVHD) – Characterized by a chronic disease course that can involve virtually all organs with variable manifestations, including sclerosis

GVHD overlap syndrome – Defined by simultaneous features of both cGVHD and aGVHD

Acute GVHD – Pathologically, aGVHD is manifest as an inflammatory immune cell infiltrate involving T cells, macrophages, monocytes and neutrophil granulocytes with associated tissue destruction and apoptosis. The transplantation conditioning regimen, innate immune system, impaired tissue repair mechanisms, and gastrointestinal microbiome all contribute to the pathophysiology of aGVHD. (See 'Acute GVHD' above.)

Chronic GVHD – Tissues affected by cGVHD are relatively acellular and fibroproliferative. The pathogenesis of cGVHD is a complex process that involves early inflammation and tissue injury, chronic inflammation and dysregulated immunity, and aberrant tissue repair and fibrosis. Description of the cellular components and signaling pathways the mediate cGVHD are discussed above. (See 'Chronic GVHD' above.)

Histocompatibility antigens – Proteins of the major histocompatibility complex (MHC) are the principal antigenic determinants of graft rejection; in humans, MHC proteins are called human leukocyte antigens (HLA). Genes of the HLA locus encode two distinct classes of cell surface molecules, class I and class II, which are expressed by different cell types, are highly polymorphic from individual to individual, and provide the surface upon which foreign antigens are displayed for immune recognition by T lymphocytes. (See 'MHC/HLA antigens' above.)

  1. Vigorito AC, Campregher PV, Storer BE, et al. Evaluation of NIH consensus criteria for classification of late acute and chronic GVHD. Blood 2009; 114:702.
  2. Zeiser R, Blazar BR. Acute Graft-versus-Host Disease - Biologic Process, Prevention, and Therapy. N Engl J Med 2017; 377:2167.
  3. Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet 2009; 373:1550.
  4. Jenq RR, Ubeda C, Taur Y, et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med 2012; 209:903.
  5. Mathewson ND, Jenq R, Mathew AV, et al. Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease. Nat Immunol 2016; 17:505.
  6. Cooke KR, Luznik L, Sarantopoulos S, et al. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 23:211.
  7. Legoff J, Resche-Rigon M, Bouquet J, et al. The eukaryotic gut virome in hematopoietic stem cell transplantation: new clues in enteric graft-versus-host disease. Nat Med 2017; 23:1080.
  8. Heidegger S, van den Brink MR, Haas T, Poeck H. The role of pattern-recognition receptors in graft-versus-host disease and graft-versus-leukemia after allogeneic stem cell transplantation. Front Immunol 2014; 5:337.
  9. Ramadan A, Paczesny S. Various forms of tissue damage and danger signals following hematopoietic stem-cell transplantation. Front Immunol 2015; 6:14.
  10. Zeiser R, Penack O, Holler E, Idzko M. Danger signals activating innate immunity in graft-versus-host disease. J Mol Med (Berl) 2011; 89:833.
  11. Blazar BR, Murphy WJ, Abedi M. Advances in graft-versus-host disease biology and therapy. Nat Rev Immunol 2012; 12:443.
  12. Wilhelm K, Ganesan J, Müller T, et al. Graft-versus-host disease is enhanced by extracellular ATP activating P2X7R. Nat Med 2010; 16:1434.
  13. Reichenbach DK, Schwarze V, Matta BM, et al. The IL-33/ST2 axis augments effector T-cell responses during acute GVHD. Blood 2015; 125:3183.
  14. Spoerl S, Mathew NR, Bscheider M, et al. Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease. Blood 2014; 123:3832.
  15. Choi J, Cooper ML, Alahmari B, et al. Pharmacologic blockade of JAK1/JAK2 reduces GvHD and preserves the graft-versus-leukemia effect. PLoS One 2014; 9:e109799.
  16. Carniti C, Gimondi S, Vendramin A, et al. Pharmacologic Inhibition of JAK1/JAK2 Signaling Reduces Experimental Murine Acute GVHD While Preserving GVT Effects. Clin Cancer Res 2015; 21:3740.
  17. Stickel N, Hanke K, Marschner D, et al. MicroRNA-146a reduces MHC-II expression via targeting JAK/STAT signaling in dendritic cells after stem cell transplantation. Leukemia 2017; 31:2732.
  18. Hülsdünker J, Ottmüller KJ, Neeff HP, et al. Neutrophils provide cellular communication between ileum and mesenteric lymph nodes at graft-versus-host disease onset. Blood 2018; 131:1858.
  19. MacDonald KP, Hill GR, Blazar BR. Chronic graft-versus-host disease: biological insights from preclinical and clinical studies. Blood 2017; 129:13.
  20. Coghill JM, Sarantopoulos S, Moran TP, et al. Effector CD4+ T cells, the cytokines they generate, and GVHD: something old and something new. Blood 2011; 117:3268.
  21. Shlomchik WD, Lee SJ, Couriel D, Pavletic SZ. Transplantation's greatest challenges: advances in chronic graft-versus-host disease. Biol Blood Marrow Transplant 2007; 13:2.
  22. Amarnath S, Flomerfelt FA, Costanzo CM, et al. Rapamycin generates anti-apoptotic human Th1/Tc1 cells via autophagy for induction of xenogeneic GVHD. Autophagy 2010; 6:523.
  23. Koehn BH, Apostolova P, Haverkamp JM, et al. GVHD-associated, inflammasome-mediated loss of function in adoptively transferred myeloid-derived suppressor cells. Blood 2015; 126:1621.
  24. Koehn BH, Saha A, McDonald-Hyman C, et al. Danger-associated extracellular ATP counters MDSC therapeutic efficacy in acute GVHD. Blood 2019; 134:1670.
  25. Rückert T, Andrieux G, Boerries M, et al. Human β-defensin 2 ameliorates acute GVHD by limiting ileal neutrophil infiltration and restraining T cell receptor signaling. Sci Transl Med 2022; 14:eabp9675.
  26. Lindemans CA, Calafiore M, Mertelsmann AM, et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature 2015; 528:560.
  27. Takashima S, Kadowaki M, Aoyama K, et al. The Wnt agonist R-spondin1 regulates systemic graft-versus-host disease by protecting intestinal stem cells. J Exp Med 2011; 208:285.
  28. Krijanovski OI, Hill GR, Cooke KR, et al. Keratinocyte growth factor separates graft-versus-leukemia effects from graft-versus-host disease. Blood 1999; 94:825.
  29. Norona J, Apostolova P, Schmidt D, et al. Glucagon-like peptide 2 for intestinal stem cell and Paneth cell repair during graft-versus-host disease in mice and humans. Blood 2020; 136:1442.
  30. Cooke KR, Gerbitz A, Crawford JM, et al. LPS antagonism reduces graft-versus-host disease and preserves graft-versus-leukemia activity after experimental bone marrow transplantation. J Clin Invest 2001; 107:1581.
  31. Zhao Y, Liu Q, Yang L, et al. TLR4 inactivation protects from graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Cell Mol Immunol 2013; 10:165.
  32. Hill GR, Teshima T, Gerbitz A, et al. Differential roles of IL-1 and TNF-alpha on graft-versus-host disease and graft versus leukemia. J Clin Invest 1999; 104:459.
  33. Jankovic D, Ganesan J, Bscheider M, et al. The Nlrp3 inflammasome regulates acute graft-versus-host disease. J Exp Med 2013; 210:1899.
  34. Holler E, Rogler G, Herfarth H, et al. Both donor and recipient NOD2/CARD15 mutations associate with transplant-related mortality and GvHD following allogeneic stem cell transplantation. Blood 2004; 104:889.
  35. MacDonald KP, Blazar BR, Hill GR. Cytokine mediators of chronic graft-versus-host disease. J Clin Invest 2017; 127:2452.
  36. Zeiser R, Nguyen VH, Beilhack A, et al. Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood 2006; 108:390.
  37. Wu T, Young JS, Johnston H, et al. Thymic damage, impaired negative selection, and development of chronic graft-versus-host disease caused by donor CD4+ and CD8+ T cells. J Immunol 2013; 191:488.
  38. Hill GR, Olver SD, Kuns RD, et al. Stem cell mobilization with G-CSF induces type 17 differentiation and promotes scleroderma. Blood 2010; 116:819.
  39. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol 2010; 10:490.
  40. Matsuoka K, Kim HT, McDonough S, et al. Altered regulatory T cell homeostasis in patients with CD4+ lymphopenia following allogeneic hematopoietic stem cell transplantation. J Clin Invest 2010; 120:1479.
  41. Sakoda Y, Hashimoto D, Asakura S, et al. Donor-derived thymic-dependent T cells cause chronic graft-versus-host disease. Blood 2007; 109:1756.
  42. Zhang P, Lee JS, Gartlan KH, et al. Eomesodermin promotes the development of type 1 regulatory T (TR1) cells. Sci Immunol 2017; 2.
  43. Highfill SL, Rodriguez PC, Zhou Q, et al. Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13. Blood 2010; 116:5738.
  44. Blazar BR, MacDonald KPA, Hill GR. Immune regulatory cell infusion for graft-versus-host disease prevention and therapy. Blood 2018; 131:2651.
  45. Patriarca F, Skert C, Sperotto A, et al. The development of autoantibodies after allogeneic stem cell transplantation is related with chronic graft-vs-host disease and immune recovery. Exp Hematol 2006; 34:389.
  46. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 2012; 18:1028.
  47. Gartlan KH, Bommiasamy H, Paz K, et al. A critical role for donor-derived IL-22 in cutaneous chronic GVHD. Am J Transplant 2018; 18:810.
  48. Alexander KA, Flynn R, Lineburg KE, et al. CSF-1-dependant donor-derived macrophages mediate chronic graft-versus-host disease. J Clin Invest 2014; 124:4266.
  49. Prasad VK, Kernan NA, Heller G, et al. DNA typing for HLA-A and HLA-B identifies disparities between patients and unrelated donors matched by HLA-A and HLA-B serology and HLA-DRB1. Blood 1999; 93:399.
  50. Schlegel PG, Aharoni R, Smilek DE, et al. Prevention of graft-versus-host disease by peptides binding to class II major histocompatibility complex molecules. Blood 1994; 84:2802.
  51. Jameson BA, McDonnell JM, Marini JC, Korngold R. A rationally designed CD4 analogue inhibits experimental allergic encephalomyelitis. Nature 1994; 368:744.
  52. Brandstadter JD, Maillard I. Notch signalling in T cell homeostasis and differentiation. Open Biol 2019; 9:190187.
  53. Socié G, Kean LS, Zeiser R, Blazar BR. Insights from integrating clinical and preclinical studies advance understanding of graft-versus-host disease. J Clin Invest 2021; 131.
  54. Holler E, Butzhammer P, Schmid K, et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol Blood Marrow Transplant 2014; 20:640.
  55. Taur Y, Xavier JB, Lipuma L, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis 2012; 55:905.
  56. Golob JL, Pergam SA, Srinivasan S, et al. Stool Microbiota at Neutrophil Recovery Is Predictive for Severe Acute Graft vs Host Disease After Hematopoietic Cell Transplantation. Clin Infect Dis 2017; 65:1984.
  57. Stoma I, Littmann ER, Peled JU, et al. Compositional Flux Within the Intestinal Microbiota and Risk for Bloodstream Infection With Gram-negative Bacteria. Clin Infect Dis 2021; 73:e4627.
  58. Peled JU, Gomes ALC, Devlin SM, et al. Microbiota as Predictor of Mortality in Allogeneic Hematopoietic-Cell Transplantation. N Engl J Med 2020; 382:822.
  59. Taur Y, Jenq RR, Perales MA, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood 2014; 124:1174.
  60. Peled JU, Devlin SM, Staffas A, et al. Intestinal Microbiota and Relapse After Hematopoietic-Cell Transplantation. J Clin Oncol 2017; 35:1650.
  61. Haak BW, Littmann ER, Chaubard JL, et al. Impact of gut colonization with butyrate-producing microbiota on respiratory viral infection following allo-HCT. Blood 2018; 131:2978.
Topic 3545 Version 30.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟