Pathogenesis of Graft-Versus-Host Disease (GVHD) - UpToDate
Pathogenesis of Graft-Versus-Host Disease (GVHD) - UpToDate
Pathogenesis of Graft-Versus-Host Disease (GVHD) - UpToDate
All topics are updated as new evidence becomes available and our peer review process is complete.
Literature review current through: Sep 2020. | This topic last updated: Apr 23, 2020.
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 is described as
acute GVHD (aGVHD) within 100 days after HCT or chronic GVHD (cGVHD) after 100 days, but
these syndromes may overlap in time.
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, diagnosis, and grading of chronic graft-
versus-host disease" and "Prevention of acute graft-versus-host disease" and "Treatment of
chronic graft-versus-host disease".)
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 [1]. Successful
transplantation requires that the donor immune system develop tolerance to these
https://www.uptodate.com/contents/pathogenesis-of-graft-versus-host-disease-gvhd/print?search=graft versus host desease&source=search_result&selectedTitl… 1/12
12/10/2020 Pathogenesis of graft-versus-host disease (GVHD) - UpToDate
alloantigens, while maintaining the ability to recognize and respond to foreign antigens,
such as microorganisms or tumor cells.
GVHD is manifest clinically as two syndromes that are generally defined temporally:
● Acute GVHD (aGVHD) – Clinical findings develop ≤100 days after HCT
● Chronic GVHD (cGVHD) – Clinical findings develop >100 days after HCT
Both aGVHD and cGVHD are consequences of the interplay between cellular/immune
mediators from the 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. As an example, aGVHD is typically manifest as an
inflammatory T cell infiltrate with tissue destruction. In contrast, the tissue response in
cGVHD is relatively acellular with fibroproliferative findings. Whereas aGVHD is primarily
driven by activation of donor T lymphocytes and release of pro-inflammatory cytokines,
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. Details of the pathophysiologic mechanisms that underlie aGVHD
and cGVHD are provided in the sections below. It remains 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
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 [4]. 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 [9-13]. 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) [14-16]. 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 [4,17]. 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 [18].
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
reduces aGVHD [19-22]. The clinical relevance of TLRs to GVHD pathophysiology is reinforced
by the observation that clinical outcomes in HCT are affected by polymorphisms of proteins
that mediate innate immunity [23]. 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, which 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 [23]. 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.
https://www.uptodate.com/contents/pathogenesis-of-graft-versus-host-disease-gvhd/print?search=graft versus host desease&source=search_result&selectedTitl… 3/12
12/10/2020 Pathogenesis of graft-versus-host disease (GVHD) - UpToDate
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. (See "Prevention of acute graft-versus-host disease",
section on 'Antibiotics'.)
Chronic GVHD — Chronic GVHD (cGVHD) is manifest as fibrosis of skin, lungs, GI tract, and
soft tissues that generally presents at least 100 days after transplantation. Clinical aspects of
cGVHD are discussed separately. (See "Clinical manifestations, diagnosis, and grading of
chronic graft-versus-host disease", section on 'Clinical and histologic features'.)
● 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 [4], as described above. (See 'Acute GVHD' above.)
Activation and injury of ECs contribute to early inflammation in cGVHD [4]. 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 [4]. 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
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 [32]. 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 (NK) cells'.)
● Aberrant tissue repair and fibrosis – Dysregulated immunity and aberrant tissue repair
contribute to scarring and fibrosis in cGVHD [4]. 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 [33]. IL-22
may also contribute to cutaneous manifestations of cGVHD [34]. 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 [4]. 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 [35].
CONTRIBUTING FACTORS
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 [36]. 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".)
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
[37,38]. 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
Clinical factors — A number of clinical variables are associated with the development of
GVHD and may influence the underlying pathophysiology [4]. 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".)
● Sex-mismatch
● Post-transplantation cyclophosphamide
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'.)
An increase of potentially pathogenic bacteria and loss of diversity in the number of bacterial
taxa is commonly found in patients undergoing allogeneic HCT [39-42]. 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
[43]. The study profiled 8767 fecal samples from 1362 patients at four institutions and used
https://www.uptodate.com/contents/pathogenesis-of-graft-versus-host-disease-gvhd/print?search=graft versus host desease&source=search_result&selectedTitl… 7/12
12/10/2020 Pathogenesis of graft-versus-host disease (GVHD) - UpToDate
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 [44-46].
SUMMARY
● 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 two syndromes that are generally defined
temporally (see 'Overview of GVHD' above):
• Acute GVHD (aGVHD) – Clinical findings develop ≤100 days after HCT
• Chronic GVHD (cGVHD) – Clinical findings develop >100 days after HCT
Cells of the innate immune system (eg, macrophages, dendritic cells, neutrophils, natural
killer [NK] cells) produce inflammatory cytokines that enhance antigen presentation and
promote differentiation of T and B lymphocytes.
● 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.)
REFERENCES
1. Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet 2009; 373:1550.
5. 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.
6. Ramadan A, Paczesny S. Various forms of tissue damage and danger signals following
hematopoietic stem-cell transplantation. Front Immunol 2015; 6:14.
8. Blazar BR, Murphy WJ, Abedi M. Advances in graft-versus-host disease biology and
therapy. Nat Rev Immunol 2012; 12:443.
9. Spoerl S, Mathew NR, Bscheider M, et al. Activity of therapeutic JAK 1/2 blockade in
graft-versus-host disease. Blood 2014; 123:3832.
10. 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.
12. 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.
13. 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.
14. MacDonald KP, Hill GR, Blazar BR. Chronic graft-versus-host disease: biological insights
from preclinical and clinical studies. Blood 2017; 129:13.
15. 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.
16. 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.
17. 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.
19. 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.
20. 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.
21. 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.
22. Jankovic D, Ganesan J, Bscheider M, et al. The Nlrp3 inflammasome regulates acute
graft-versus-host disease. J Exp Med 2013; 210:1899.
23. 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.
24. MacDonald KP, Blazar BR, Hill GR. Cytokine mediators of chronic graft-versus-host
disease. J Clin Invest 2017; 127:2452.
25. 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.
26. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the
human immune system. Nat Rev Immunol 2010; 10:490.
27. 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.
29. Zhang P, Lee JS, Gartlan KH, et al. Eomesodermin promotes the development of type 1
regulatory T (TR1) cells. Sci Immunol 2017; 2.
30. 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.
31. Blazar BR, MacDonald KPA, Hill GR. Immune regulatory cell infusion for graft-versus-
host disease prevention and therapy. Blood 2018; 131:2651.
33. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic
disease. Nat Med 2012; 18:1028.
34. 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.
35. 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.
https://www.uptodate.com/contents/pathogenesis-of-graft-versus-host-disease-gvhd/print?search=graft versus host desease&source=search_result&selectedTit… 11/12
12/10/2020 Pathogenesis of graft-versus-host disease (GVHD) - UpToDate
36. 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.
37. 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.
38. Jameson BA, McDonnell JM, Marini JC, Korngold R. A rationally designed CD4 analogue
inhibits experimental allergic encephalomyelitis. Nature 1994; 368:744.
39. 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.
40. 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.
41. 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.
42. 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 2020.
43. 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.
44. 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.
45. Peled JU, Devlin SM, Staffas A, et al. Intestinal Microbiota and Relapse After
Hematopoietic-Cell Transplantation. J Clin Oncol 2017; 35:1650.
46. 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.