Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jul 15.
Published in final edited form as: Biol Blood Marrow Transplant. 2008 Jan;15(1 Suppl):e1–e7. doi: 10.1016/j.bbmt.2008.12.500

Genomic and Proteomic Analysis of Allogeneic Hematopoietic Cell Transplant Outcome. Seeking Greater Understanding the Pathogenesis of GVHD and Mortality

John A Hansen 1
PMCID: PMC2710961  NIHMSID: NIHMS89444  PMID: 19147066

Success following allogeneic hematopoietic cell transplantation (HSCT) is ultimately determined by the ability to achieve sustained engraftment and immune reconstitution, eradication of the abnormal or malignant cells responsible for the patient’s disease, and control of graft-versus-host disease (GVHD). GVHD, an immune mediated reaction initiated by donor T cells in response to host alloantigen is the cause of significant morbidity and death in many patients. Genetic matching for HLA, the human major histocompatibility complex (MHC) located on chromosome 6p21 has for several years become well established as a requirement for optimal HSCT outcome [1]. Despite complete matching for all variation spanning 4Mb of DNA across the MHC, acute GVHD, chronic GVHD and transplant related mortality (TRM) occurs in a significant number of HLA identical sibling donor transplants.

The alloimmune reaction

Clinical GVHD results from an alloimmune reaction that occurs when immune competent donor T cells are transplanted to an immune compromised host and the genetic differences between donor and recipient are sufficient to induce T cell activation. [2] The genetic differences responsible for an allograft reaction encode polymorphic cellular proteins called histocompatibility antigens. [3] The strongest histocompatibility antigens are encoded by the class I and class II genes of the major histocompatibility complex (MHC), or HLA, but there are many other genes located throughout the genome that encode cellular peptides capable of generating significant alloimmune responses if variation in the gene product can be detected by T cells. These “allo” peptides are called minor histocompatibility antigens (mHA). [4] Although HLA identical siblings share 50% of their genome, the mHA disparity is sufficient to cause clinically significant acute GVHD in 30–40% of cases. Despite HLA matching among unrelated donor-recipient pairs, disparity for non-MHC mHA is greater because the recipient and donor do not share the same parental chromosomes. [5]

Pathogenesis of GVHD

The incidence, severity and duration of GVHD following HSCT vary substantially from patient to patient. [611] Differences in GVHD phenotype can result from both inherited and clinical or environmental factors including type of disease, disease status, treatment history, HLA match, age, sex of donor and patient (and sex match), and the type of conditioning therapy administered prior to transplant. [8,12,13]

High-dose cytotoxic conditioning can increase the risk of acute GVHD by causing mucosal injury which facilitates translocation of endotoxin into the bloodstream, suppressing T cell and NK cell mediated allograft resistance and activating antigen-presenting cells in the recipient. Within hours after transplantation, GVHD begins with the presentation of alloantigens by host dendritic cells, followed by activation of donor T cells which undergo clonal expansion and migration into various lymphoid organs and target tissues. [14] The anti-host alloimmune reaction is largely driven by the leakage of lipopolysaccharide (LPS) across the gut wall, [15] and further amplified by multiple components of the immune systems, including NK cells, monocytes, macrophages and proinflammatory cytokines. [1620]

Proteomic analysis of acute GVHD and transplant-related mortality

Several studies have described changes in plasma proteins that correlate with the risk of acute GVHD, chronic GVHD and TRM (summarized in Table 1). Although there are common discoveries among several of these studies, there are also findings that have not independently replicated and for this reason true positives have not been clearly defined. There remains a great need for further and rigorously conducted studies that achieve both validation and also identification of the clinical covariables that in must be known to fully interpret and reliably implement biomarker data, both proteomic and genomic, into clinical risk assessment algorithms and pre-emptive therapies.

Table 1.

Summary of published changes in plasma proteins associated with acute GVHD, TRM and chronic GVHD. 1

Protein aGVHD TRM cGVHD
IL1-RN Liem 1998 [54]
IL2R Miyamoto 1996 [55]; Grimm 1998 [56]; Foley 1998 [57]; Nakamura 2000 [58]; Visentainer 2003 [59]; Shaiegan 2006 [60]; Paczesny 2008 [61] Liem 1998 [54]; Fujii 2008 [62]
IL-6 Imamura 1994 [63]
IL-8 Uguccioni 1993 [64]; Paczesny 2008 [61] Schots 2003 [65]
IL-10 Liem 1998 [54] Liem 1998 [54]; Visentainer 2003 [59]
IL-12 Nakamura 2000 [58]; Mohty 2005 [66]
IL-15 Sakata 2001 [67]
IL-18 Nakamura 2000 [58]; Fujimori 2000 [68]; Shaiegan 2006 [60]; Luft 2007 [69]
BAFF Fujii 2008 [62]
CD13 Fujii 2008 [62]
CCL8 Hori 2008 [70]
CXCL10 Piper 2007 [71]
HGF Okamoto 2001 [72]; Paczesny 2008 [61]
IFNG Imamura 1994 [63] ; Nakamura 2000 [58]
TNF Holler 1990 [73]; Symington 1990 [74]; Imamura 1994 [63]
TNFR Or 1996 [75]; Kitko 2008 [76]; Choi 2008 [77]; Paczesny 2008 [61]
Syndecan-1 Seidel 2003 [78]
anti-dsDNA Fujii 2008 [62]
1

Refer to original publication for additional details.

Genomic analysis GVHD and TRM

Several studies over the last 10 years have identified genetic polymorphisms associated with GVHD and TRM (summarized in Table 2). Initially these investigations focused on well known genes encoding proinflammatory or immune modulating cytokines including IL1A, IL1B, IL1RN, IL6, IL10, INFG, TGFB and TNF. [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] Subsequent studies have examined additional genes for variation associated with HSCT outcomes including CTLA4, ESR1, IL2, IL7R, IL8, IL10RB, IL18, NOD2, VDR. [28,35,36] [37] [38] [39] [40] [41] [42] [43]

Table 2.

Genetic variation in immune response genes associated with acute GVHD, TRM and chronic GVHD.

Gene HSCT associated phenotype Study population (N) 1 VNTR or SNP Discovery and supporting reference(s) 3 Investigators reporting non-significant results 3
alias location alleles rs number
CTLA4 (CD152) aGVHD & survival MRD (536) +49 (d), CT60 (d) 2 A/G, G/A rs231775, rs3087243 Perez-Garcia 2007 [41]
cGVHD +49/GG A/G rs231775 Azarian 2007 [42]
ESR1 aGVHD MRD (108) VNTR, PvuII & XbaI intron 1 - - - - Middleton 2003 [35]
FAS aGVHD MRD (160) −670 (p) promoter A/G - - Mullighan 2004 [29]
IFNG aGVHD MRD (49) VNTR intron 1 na - - Middleton 1998 [21]
aGVHD MRD (100) 874 T/A - - - - Socie 2001 [23]
IL1A cGVHD MRD (115) −899, VNTR promoter, intron 6 C/T, allele 2 - -
- -
Cullup 2003 [27] - -
aGVHD & TRM URD( 426) −889 promoter C/T - - - - Mehta 2007 [34]
IL1B aGVHD MRD −511 (p) promoter T/C rs16944 MacMillan BJH 2003[28] - -
aGVHD MRD (570) −511 (p/d) promoter C/T rs16944 - - Lin 2003 [26]
IL1RN aGVHD MRD (99) VNTR intron 1 - - na Cullup 2001 [24] - -
aGVHD & cGVHD MRD (107) VNTR (d) intron 2 absence of allele *2 - - Rocha 2002 [25] - -
MRD (570) 9261 (p/d) intron 1 G/A 448341 - - Lin 2003 [26]
IL2 aGVHD URD (95) −333 (p) promoter T/G rs2069762 MacMillan Transplant 2003 [43] - -
IL6 aGVHD MRD (160) −174 (d) promoter G/C rs1800795 Mullighan 2004 [29] - -
aGVHD ND (93) −174 (p/d) promoter G/C rs1800795 Karabon 2005 [31] - -
cGVHD −174 promoter G/C rs1800795 Cavet 1999 [22] - -
cGVHD MRD (100) −174 (p) promoter G/C Socie 2001 [23] - -
aGVHD, cGVHD, TRM MRD (570) −174 (d) promoter G/C rs1800795 - - Lin 2003 [26]
IL7R TRM MURD (75) +1237(p) cSNP A/G - - Shamim 2006 [40] - -
TRM MRD (100) +510,+1237, +2087,+3101 cSNPs C/T,A/G,C/T,A/G - - - - Shamim 2006 [40]
IL10 aGVHD MRD (49) IL10G; 4 promoter - - - - Middleton 1998 [21] - -
aGVHD MRD (144) IL10/−1082, IL10−1064 (4) promoter A/G - - Cavet 1999 [22] - -
aGVHD MRD (100) hap 5 promoter G-C-C - - Socie 2001 [23] - -
aGVHD MRD (993) −592 (p) & hap (p) 5 promoter C/A, A-T-A Lin 2003 [26] - -
aGVHD MRD (160) ATA hap(p) promoter - - Mullighan 2004 [29]
cGVHD MRD (107) 1082*G/G(p) promoter A/G - - Rocha 2002 [25] - -
cGVHD ATA hap(p) promoter Mullighan 2004 [29] - -
TRM URD (182) IL10R24-SNP hap (d) 5 promoter G-C-C Keen 2004 [30] - -
IL10RB aGVHD MRD (993) c238 (d) exon Lin 2005 [37] - -
IL18 Survival URD (157) GCG hap(p) Cardodo 2004 [38] - -
NOD2 aGVHD & TRM SNP8, 12, 3 intragenic G/A, G/C, insertion rs2066844, rs2066845, rs2066847 Holler 2004, 2006 [36,39] - -
TNF aGVHD MRD (49) TNFd 6,7 VNTR na Middleton 1998 [21]
TRM MRD (144) TNFd 7 VNTR na Cavet 1999 [22]
aGVHD MRD (100) MRD ( 570) MRD (160) −308 7 promoter G/A rs1800629 - - Socie 2001 [23]; Lin 2003 [26]; Mullighan 2004 [29]
TRM URD (182) TNFd & −1031 promoter: VNTR & SNP TNFd*4/−1031*C hap - - Keen 2004 [30] - -
aGVHD & TRM MRD (160) 488 7 Mullighan 2004 [29] - -
TNFRII cGVHD MRD (104) codon 196 exon 6 T/G Stark 2003 [79] - -
VDR aGVHD & TRM MRD (88) VNTR (d) intron 8 Middleton 2002 [80] - -
1

Study population: “MRD”, indicates HLA matched related donor; “ND”, not defined; “URD”, unrelated donor.

2

“p”, indicates patient; “d”, donor.

3

Refer to original publication(s) for additional details.

4

IL10G and IL10R are a microsatellites located in and nearby the IL10 gene.

5

IL10 promoter region haplotype, positions: −1082/−819/−592

6

TNFd is a microsatellite in the TBF region.

7

MRD cases, because they are HLA identical, share the same TNF genotypes

Unfortunately, the results of most of these single Center studies have not been independently validated by others in separate patient populations. Lack of validation or inconsistency in these results may be due largely to lack of statistical power since many of the original studies were based on relatively small numbers (<200–300 cases). Nevertheless, these results have been sufficiently compelling to warrant additional study. Comprehensive critical reviews of this research have been recently published [44],[45] and other current papers have addressed the potential impact of developments in genomic sciences and the opportunity for expanding HSCT outcomes research to genome-wide discovery. [1,46]

Genome-wide association studies (GWAS)

The remarkable development in recent years of methods and tools for the characterization of the entire human genome has dramatically broadened the opportunity for the genetic analysis of disease. The recent completion of the human genome map[47,48] and the development of dense SNP marker maps of the genome, [49,50] as well as development of massively parallel genotyping technologies, [5153] have made it possible to screen genes in an unbiased manner for polymorphisms that correlate with any well defined phenotype, disease status or relevant quantitative trait. This is particularly important when considering complex traits that characterize HCT complications and outcomes. Consideration of the entire genome in an unbiased fashion permits the discovery of genetic factors that would have never been considered otherwise.

State of the art genomic and proteomic studies of GVHD and mortality

The three papers that follow this Introduction are summaries of the oral presentations that will be given during the Genomics and Proteomics Scientific Session of the 2009 BMT Tandem meetings. These papers are each timely progress reports of our emerging understanding of the pathogenesis of GVHD and TRM. The paper by Sophie Paczesny, Jamie Ferrara and team provides model example of the rigorous two phase, discovery and validation, proteomic study of changes in plasma proteins associated with the development of acute GVHD. The technology used for the discovery phase used an antibody array containing antibodies specific for 120 human proteins including acute phase reactants, cytokines, angiogenic factors, tumor markers, leukocyte adhesion molecules and metalloproteinases and their inhibitors. The papers by Seishi Ogawa et al and Jason Chien et al describe preliminary discovery data from two of the first large whole genome scans performed on DNA from both recipient and donor as a comprehensive approach to examining genetic disparity and GVHD (Ogawa et al), and the association of genetic variation with transplant outcomes including gram-negative bacteremia and bronchiolitis obliterans.

Acknowledgments

This work was supported by grants from the National Institutes of Health AI33484, CA015704, CA18029, HL087690 and HL094260.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Hansen JA, Petersdorf EW, Lin MT, et al. Genetics of allogeneic hematopoietic cell transplantation. Role of HLA matching, functional variation in immune response genes. Immunol Res. 2008;41:56–78. doi: 10.1007/s12026-007-0043-x. [DOI] [PubMed] [Google Scholar]
  • 2.Elkins WL. Cellular immunology and the pathogenesis of graft versus host reactions (Review) Prog Allergy. 1971;15:78–187. [PubMed] [Google Scholar]
  • 3.Snell GD, Dausset J, Nathenson S. Histocompatibility. New York, NY: Academic Press; 1976. [Google Scholar]
  • 4.Perreault C, Décary F, Brochu S, Gyger M, Bélanger R, Roy D. Minor histocompatibility antigens. Blood. 1990;76:1269–1280. [PubMed] [Google Scholar]
  • 5.Martin PJ. Increased disparity for minor histocompatibility antigens as a potential cause of increased GVHD risk in marrow transplantation from unrelated donors compared with related donors. Bone Marrow Transplant. 1991;8:217–223. [PubMed] [Google Scholar]
  • 6.Glucksberg H, Storb R, Fefer A, et al. Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors. Transplantation. 1974;18:295–304. doi: 10.1097/00007890-197410000-00001. [DOI] [PubMed] [Google Scholar]
  • 7.Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2005;11:945–956. doi: 10.1016/j.bbmt.2005.09.004. [DOI] [PubMed] [Google Scholar]
  • 8.Nash RA, Pepe MS, Storb R, et al. Acute graft-versus-host disease: analysis of risk factors after allogeneic marrow transplantation and prophylaxis with cyclosporine and methotrexate. Blood. 1992;80:1838–1845. [PubMed] [Google Scholar]
  • 9.Przepiorka D, Weisdorf D, Martin P, et al. 1994 Consensus conference on acute GVHD grading. Bone Marrow Transplant. 1995;15:825–828. [PubMed] [Google Scholar]
  • 10.Stewart BL, Storer B, Storek J, et al. Duration of immunosuppressive treatment for chronic graft-versus-host disease. Blood. 2004;104:3501–3506. doi: 10.1182/blood-2004-01-0200. [DOI] [PubMed] [Google Scholar]
  • 11.Martin PJ, McDonald GB, Sanders JE, et al. Increasingly frequent diagnosis of acute gastrointestinal graft-versus-host disease after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2004;10:320–327. doi: 10.1016/j.bbmt.2003.12.304. [DOI] [PubMed] [Google Scholar]
  • 12.Przepiorka D, Smith TL, Folloder J, et al. Risk factors for acute graft-versus-host disese after allogeneic blood stem cell transplantation. Blood. 1999;94:1465–1470. [PubMed] [Google Scholar]
  • 13.Mielcarek M, Martin PJ, Leisenring W, et al. Graft-versus-host disease after nonmyeloablative versus conventional hematopoietic stem cell transplantation. Blood. 2003;102:756–762. doi: 10.1182/blood-2002-08-2628. [DOI] [PubMed] [Google Scholar]
  • 14.Beilhack A, Schulz S, Baker J, et al. In vivo analyses of early events in acute graft-versus-host disease reveal sequential infiltration of T-cell subsets. Blood. 2005;106:1113–1122. doi: 10.1182/blood-2005-02-0509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Nestel FP, Price KS, Seemayer TA, Lapp WS. Macrophage priming and lipopolysaccharide-triggered release of tumor necrosis factor α during graft-versus-host disease. J Exp Med. 1992;175:405–413. doi: 10.1084/jem.175.2.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ferrara JLM, Deeg HJ. Graft-versus-host disease (Review) N Engl J Med. 1991;324:667–674. doi: 10.1056/NEJM199103073241005. [DOI] [PubMed] [Google Scholar]
  • 17.Antin JH, Ferrara JLM. Cytokine dysregulation and acute graft-versus-host disease. Blood. 1992;80:2964–2968. [PubMed] [Google Scholar]
  • 18.Jadus MR, Websic HT. The role of cytokines in graft-versus-host reactions and disease. Bone Marrow Transplant. 1992;10:1–14. [PubMed] [Google Scholar]
  • 19.Holler E, Kolb HJ, Mittermuller J, et al. Modulation of acute graft-versus-host-disease after allogeneic bone marrow transplantation by tumor necrosis factor alpha (TNF alpha) release in the course of pretransplant conditioning: role of conditioning regimens and prophylactic application of a monoclonal antibody neutralizing human TNF alpha (MAK 195F) Blood. 1995;86:890–899. [PubMed] [Google Scholar]
  • 20.Krenger W, Hill GR, Ferrara JL. Cytokine cascades in acute graft-versus-host disease (Review) Transplantation. 1997;64:553–558. doi: 10.1097/00007890-199708270-00001. [DOI] [PubMed] [Google Scholar]
  • 21.Middleton PG, Taylor PRA, Jackson G, Proctor SJ, Dickinson AM. Cytokine gene polymorphisms associating with severe acute graft-versus-host disease in HLA-identical sibling transplants. Blood. 1998;92:3943–3948. [PubMed] [Google Scholar]
  • 22.Cavet J, Middleton PG, Segall M, Noreen H, Davies SM, Dickinson AM. Recipient tumor necrosis factor-alpha and interleukin-10 gene polymorphisms associate with early mortality and acute graft-versus-host disease severity in HLA-matched sibling bone marrow transplants. Blood. 1999;94:3941–3946. [PubMed] [Google Scholar]
  • 23.Socie G, Loiseau P, Tamouza R, et al. Both genetic and clinical factors predict the development of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Transplantation. 2001;72:699–706. doi: 10.1097/00007890-200108270-00024. [DOI] [PubMed] [Google Scholar]
  • 24.Cullup H, Dickinson AM, Jackson GH, Taylor PR, Cavet J, Middleton PG. Donor interleukin 1 receptor antagonist genotype associated with acute graft-versus-host disease in human leucocyte antigen-matched sibling allogeneic transplants. Br J Haematol. 2001;113:807–813. doi: 10.1046/j.1365-2141.2001.02811.x. [DOI] [PubMed] [Google Scholar]
  • 25.Rocha V, Franco RF, Porcher R, et al. Host defense and inflammatory gene polymorphisms are associated with outcomes after HLA-identical sibling bone marrow transplantation. Blood. 2002;100:3908–3918. doi: 10.1182/blood-2002-04-1033. [DOI] [PubMed] [Google Scholar]
  • 26.Lin M-T, Storer B, Martin PJ, et al. Relation of an interleukin-10 promoter polymorphism to graft-versus-host disease and survival after hematopoietic-cell transplantation. N Engl J Med. 2003;349:2201–2210. doi: 10.1056/NEJMoa022060. [DOI] [PubMed] [Google Scholar]
  • 27.Cullup H, Dickinson AM, Cavet J, Jackson GH, Middleton PG. Polymorphisms of interleukin-1alpha constitute independent risk factors for chronic graft-versus-host disease after allogeneic bone marrow transplantation. Br J Haematol. 2003;122:778–787. doi: 10.1046/j.1365-2141.2003.04510.x. [DOI] [PubMed] [Google Scholar]
  • 28.MacMillan ML, Radloff GA, Defor TE, Weisdorf DJ, Davies SM. Interleukin-1 genotype and outcome of unrelated donor bone marrow transplantation. Br J Haematol. 2003;121:597–604. doi: 10.1046/j.1365-2141.2003.04314.x. [DOI] [PubMed] [Google Scholar]
  • 29.Mullighan C, Heatley S, Doherty K, et al. Non-HLA immunogenetic polymorphisms and the risk of complications after allogeneic hemopoietic stem-cell transplantation. Transplantation. 2004;27:587–596. doi: 10.1097/01.tp.0000111769.45088.a2. [DOI] [PubMed] [Google Scholar]
  • 30.Keen LJ, Defor TE, Bidwell JL, Davies SM, Bradley BA, Hows JM. Interleukin-10 and tumor necrosis factor alpha region haplotypes predict transplant-related mortality after unrelated donor stem cell transplantation. Blood. 2004;103:3599–3602. doi: 10.1182/blood-2002-11-3568. [DOI] [PubMed] [Google Scholar]
  • 31.Karabon L, Wysoczanska B, Bogunia-Kubik K, Suchnicki K, Lange A. IL-6 and IL-10 promoter gene polymorphisms of patients and donors of allogeneic sibling hematopoietic stem cell transplants associate with the risk of acute graft-versus-host disease. Hum Immunol. 2005;66:700–710. doi: 10.1016/j.humimm.2005.02.003. [DOI] [PubMed] [Google Scholar]
  • 32.Seo KW, Kim DH, Sohn SK, et al. Protective role of interleukin-10 promoter gene polymorphism in the pathogenesis of invasive pulmonary aspergillosis after allogeneic stem cell transplantation. Bone Marrow Transplant. 2005;36:1089–1095. doi: 10.1038/sj.bmt.1705181. [DOI] [PubMed] [Google Scholar]
  • 33.Bogunia-Kubik K, Mlynarczewska A, Jaskula E, Lange A. The presence of IFNG 3/3 genotype in the recipient associates with increased risk for Epstein-Barr virus reactivation after allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2006;132:326–332. doi: 10.1111/j.1365-2141.2005.05875.x. [DOI] [PubMed] [Google Scholar]
  • 34.Mehta PA, Eapen M, Klein JP, et al. Interleukin-1 alpha genotype and outcome of unrelated donor haematopoietic stem cell transplantation for chronic myeloid leukaemia. Br J Haematol. 2007;137:152–157. doi: 10.1111/j.1365-2141.2007.06552.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Middleton PG, Norden J, Cullup H, et al. Oestrogen receptor alpha gene polymorphism associates with occurrence of graft-versus-host disease and reduced survival in HLA-matched sib-allo BMT. Bone Marrow Transplant. 2003;32:41–47. doi: 10.1038/sj.bmt.1704090. [DOI] [PubMed] [Google Scholar]
  • 36.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;104:889–894. doi: 10.1182/blood-2003-10-3543. [DOI] [PubMed] [Google Scholar]
  • 37.Lin M-T, Storer B, Martin PJ, et al. Genetic variation in the IL-10 pathway modulates severity of acute graft-versus-host disease following hematopoietic cell transplantation: synergism between IL-10 genotype of patient and IL-10 receptor β genotype of donor. Blood. 2005;106:3995–4001. doi: 10.1182/blood-2004-11-4338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Cardoso SM, Defor TE, Tilley LA, Bidwell JL, Weisdorf DJ, MacMillan ML. Patient interleukin-18 GCG haplotype associates with improved survival and decreased transplant-related mortality after unrelated-donor bone marrow transplantation. Br J Haematol. 2004;126:704–710. doi: 10.1111/j.1365-2141.2004.05128.x. [DOI] [PubMed] [Google Scholar]
  • 39.Holler E, Rogler G, Brenmoehl J, et al. Prognostic significance of NOD2/CARD15 variants in HLA-identical sibling hematopoietic stem cell transplantation: Effect on long term outcome is confirmed in 2 independent cohorts and may be modulated by the type of gastrointestinal decontamination. Blood. 2006;107:4189–4193. doi: 10.1182/blood-2005-09-3741. [DOI] [PubMed] [Google Scholar]
  • 40.Shamim Z, Ryder LP, Heilmann C, et al. Genetic polymorphisms in the genes encoding human interleukin-7 receptor-alpha: prognostic significance in allogeneic stem cell transplantation. Bone Marrow Transplant. 2006;37:485–491. doi: 10.1038/sj.bmt.1705277. [DOI] [PubMed] [Google Scholar]
  • 41.Perez-Garcia A, de la CR, Roman-Gomez J, et al. CTLA-4 polymorphisms and clinical outcome after allogeneic stem cell transplantation from HLA-identical sibling donors. Blood. 2007;110:461–467. doi: 10.1182/blood-2007-01-069781. [DOI] [PubMed] [Google Scholar]
  • 42.Azarian M, Busson M, Lepage V, et al. Donor CTLA-4 +49 A/G*GG genotype is associated with chronic GVHD after HLA-identical haematopoietic stem-cell transplantations. Blood. 2007;110:4623–4624. doi: 10.1182/blood-2007-08-106385. [DOI] [PubMed] [Google Scholar]
  • 43.MacMillan ML, Radloff GA, Kiffmeyer WR, Defor TE, Weisdorf DJ, Davies SM. High-producer interleukin-2 genotype increases risk for acute graft-versus-host disease after unrelated donor bone marrow transplantation. Transplantation. 2003;76:1758–1762. doi: 10.1097/01.TP.0000095899.54052.89. [DOI] [PubMed] [Google Scholar]
  • 44.Dickinson AM, Harrold JL, Cullup H. Haematopoietic stem cell transplantation: can our genes predict clinical outcome? (Review) Expert Reviews in Molecular Medicine. 2007;9:1–19. doi: 10.1017/S1462399407000488. [DOI] [PubMed] [Google Scholar]
  • 45.Mullighan CG, Bardy PG. New directions in the genomics of allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2007;13:127–144. doi: 10.1016/j.bbmt.2006.10.018. [DOI] [PubMed] [Google Scholar]
  • 46.Mullally A, Ritz J. Beyond HLA: the significance of genomic variation for allogeneic hematopoietic stem cell transplantation. Blood. 2007;109:1355–1362. doi: 10.1182/blood-2006-06-030858. [DOI] [PubMed] [Google Scholar]
  • 47.Lander ES, Linton LM, Birren B, et al. Initial sequencing and analysis of the human genome. Nature. 2001;409:860–921. doi: 10.1038/35057062. [DOI] [PubMed] [Google Scholar]
  • 48.Venter JC, Adams MD, Myers EW, et al. The sequence of the human genome. Science. 2001;291:1304–1351. doi: 10.1126/science.1058040. [DOI] [PubMed] [Google Scholar]
  • 49.Miller RD, Phillips MS, Jo I, et al. High-density single-nucleotide polymorphism maps of the human genome. Genomics. 2005;86:117–126. doi: 10.1016/j.ygeno.2005.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gabriel SB, Schaffner SF, Nguyen H, et al. The structure of haplotype blocks in the human genome. Science. 2002;296:2225–2229. doi: 10.1126/science.1069424. [DOI] [PubMed] [Google Scholar]
  • 51.Matsuzaki H, Dong S, Loi H, et al. Genotyping over 100,000 SNPs on a pair of oligonucleotide arrays. Nature Methods. 2004;1:109–111. doi: 10.1038/nmeth718. [DOI] [PubMed] [Google Scholar]
  • 52.Steemers FJ, Chang W, Lee G, Barker DL, Shen R, Gunderson KL. Whole-genome genotyping with the single-base extension assay. Nature Methods. 2006;3:31–33. doi: 10.1038/nmeth842. [DOI] [PubMed] [Google Scholar]
  • 53.Gunderson KL, Steemers FJ, Lee G, Mendoza LG, Chee MS. A genome-wide scalable SNP genotyping assay using microarray technology. Nat Genet. 2005;37:549–554. doi: 10.1038/ng1547. [DOI] [PubMed] [Google Scholar]
  • 54.Liem LM, van Houwelingen HC, Goulmy E. Serum cytokine levels after HLA-identical bone marrow transplantation. Transplantation. 1998;66:863–871. doi: 10.1097/00007890-199810150-00009. [DOI] [PubMed] [Google Scholar]
  • 55.Miyamoto T, Akashi K, Hayashi S, et al. Serum concentration of the soluble interleukin-2 receptor for monitoring acute graft-versus-host disease. Bone Marrow Transplant. 1996;17:185–190. [PubMed] [Google Scholar]
  • 56.Grimm J, Zeller W, Zander AR. Soluble interleukin-2 receptor serum levels after allogeneic bone marrow transplantations as a marker for GVHD. Bone Marrow Transplant. 1998;21:29–32. doi: 10.1038/sj.bmt.1701041. [DOI] [PubMed] [Google Scholar]
  • 57.Foley R, Couban S, Walker I, et al. Monitoring soluble interleukin-2 receptor levels in related and unrelated donor allogenic bone marrow transplantation. Bone Marrow Transplant. 1998;21:769–773. doi: 10.1038/sj.bmt.1701163. [DOI] [PubMed] [Google Scholar]
  • 58.Nakamura H, Komatsu K, Ayaki M, et al. Serum levels of soluble IL-2 receptor, IL-12, IL-18, and IFN-gamma in patients with acute graft-versus-host disease after allogeneic bone marrow transplantation. J Allergy Clin Immunol. 2000;106:S45–S50. doi: 10.1067/mai.2000.106774. [DOI] [PubMed] [Google Scholar]
  • 59.Visentainer JE, Lieber SR, Persoli LB, et al. Serum cytokine levels and acute graft-versus-host disease after HLA-identical hematopoietic stem cell transplantation. Exp Hematol. 2003;31:1044–1050. doi: 10.1016/j.exphem.2003.08.005. [DOI] [PubMed] [Google Scholar]
  • 60.Shaiegan M, Iravani M, Babaee GR, Ghavamzadeh A. Effect of IL-18 and sIL2R on aGVHD occurrence after hematopoietic stem cell transplantation in some Iranian patients. Transpl Immunol. 2006;15:223–227. doi: 10.1016/j.trim.2005.10.002. [DOI] [PubMed] [Google Scholar]
  • 61.Paczesny S, Krijanovski OI, Braun TM, et al. A biomarker panel for acute graft versus host disease. Blood. doi: 10.1182/blood-2008-07-167098. 9999;prepublished online October 2, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Fujii H, Cuvelier G, She K, et al. Biomarkers in newly diagnosed pediatric-extensive chronic graft-versus-host disease: a report from the Children’s Oncology Group. Blood. 2008;111:3276–3285. doi: 10.1182/blood-2007-08-106286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Imamura M, Hashino S, Kobayashi H, et al. Serum cytokine levels in bone marrow transplantation: synergistic interaction of interleukin-6, interferon-gamma, and tumor necrosis factor-alpha in graft-versus-host disease. Bone Marrow Transplant. 1994;13:745–751. [PubMed] [Google Scholar]
  • 64.Uguccioni M, Meliconi R, Nesci S, et al. Elevated interleukin-8 serum concentrations in beta-thalassemia and graft-versus-host disease. Blood. 1993;81:2252–2256. [PubMed] [Google Scholar]
  • 65.Schots R, Kaufman L, Van RI, et al. Proinflammatory cytokines and their role in the development of major transplant-related complications in the early phase after allogeneic bone marrow transplantation. Leukemia. 2003;17:1150–1156. doi: 10.1038/sj.leu.2402946. [DOI] [PubMed] [Google Scholar]
  • 66.Mohty M, Blaise D, Faucher C, et al. Inflammatory cytokines and acute graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation. Blood. 2005;106:4407–4411. doi: 10.1182/blood-2005-07-2919. [DOI] [PubMed] [Google Scholar]
  • 67.Sakata N, Yasui M, Okamura T, Inoue M, Yumura-Yagi K, Kawa K. Kinetics of plasma cytokines after hematopoietic stem cell transplantation from unrelated donors: the ratio of plasma IL-10/sTNFR level as a potential prognostic marker in severe acute graft-versus-host disease. Bone Marrow Transplant. 2001;27:1153–1161. doi: 10.1038/sj.bmt.1703060. [DOI] [PubMed] [Google Scholar]
  • 68.Fujimori Y, Takatsuka H, Takemoto Y, et al. Elevated interleukin (IL)-18 levels during acute graft-versus-host disease after allogeneic bone marrow transplantation. Br J Haematol. 2000;109:652–657. doi: 10.1046/j.1365-2141.2000.02095.x. [DOI] [PubMed] [Google Scholar]
  • 69.Luft T, Conzelmann M, Benner A, et al. Serum cytokeratin-18 fragments as quantitative markers of epithelial apoptosis in liver and intestinal graft-versus-host disease. Blood. 2007;110:4535–4542. doi: 10.1182/blood-2006-10-049817. [DOI] [PubMed] [Google Scholar]
  • 70.Hori T, Naishiro Y, Sohma H, et al. CCL8 is a potential molecular candidate for the diagnosis of graft-versus-host disease. Blood. 2008;111:4403–4412. doi: 10.1182/blood-2007-06-097287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Piper KP, Horlock C, Curnow SJ, et al. CXCL10-CXCR3 interactions play an important role in the pathogenesis of acute graft-versus-host disease in the skin following allogeneic stem-cell transplantation. Blood. 2007;110:3827–3832. doi: 10.1182/blood-2006-12-061408. [DOI] [PubMed] [Google Scholar]
  • 72.Okamoto T, Takatsuka H, Fujimori Y, Wada H, Iwasaki T, Kakishita E. Increased hepatocyte growth factor in serum in acute graft-versus-host disease. Bone Marrow Transplant. 2001;28:197–200. doi: 10.1038/sj.bmt.1703095. [DOI] [PubMed] [Google Scholar]
  • 73.Holler E, Kolb HJ, Möller A, et al. Increased serum levels of tumor necrosis factor α precede major complications of bone marrow transplantation. Blood. 1990;75:1011–1016. [PubMed] [Google Scholar]
  • 74.Smith JH, Rotterdam HZ, Christie JD, et al. Specialty conference on infectious and inflammatory diseases. Mod Pathol. 1990;3:223–233. [PubMed] [Google Scholar]
  • 75.Or R, Kalinkovich A, Nagler A, et al. Soluble tumor necrosis factor (sTNF) receptors: a possible prognostic marker for bone marrow transplantation-related complications. Cytokines and Molecular Therapy. 1996;2:243–250. [PubMed] [Google Scholar]
  • 76.Kitko CL, Paczesny S, Yanik G, et al. Plasma elevations of tumor necrosis factor-receptor-1 at day 7 postallogeneic transplant correlate with graft-versus-host disease severity and overall survival in pediatric patients. Biol Blood Marrow Transplant. 2008;14:759–765. doi: 10.1016/j.bbmt.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Choi SW, Kitko CL, Braun T, et al. Change in plasma tumor necrosis factor receptor 1 levels in the first week after myeloablative allogeneic transplantation correlates with severity and incidence of GVHD and survival. Blood. 2008;112:1539–1542. doi: 10.1182/blood-2008-02-138867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Seidel C, Ringden O, Remberger M. Increased levels of syndecan-1 in serum during acute graft-versus-host disease. Transplantation. 2003;76:423–426. doi: 10.1097/01.TP.0000074316.76104.A5. [DOI] [PubMed] [Google Scholar]
  • 79.Stark GL, Dickinson AM, Jackson GH, Taylor PR, Proctor SJ, Middleton PG. Tumour necrosis factor receptor type II 196M/R genotype correlates with circulating soluble receptor levels in normal subjects and with graft-versus-host disease after sibling allogeneic bone marrow transplantation. Transplantation. 2003;76:1742–1749. doi: 10.1097/01.TP.0000092496.05951.D5. [DOI] [PubMed] [Google Scholar]
  • 80.Middleton PG, Cullup H, Dickinson AM, et al. Vitamin D receptor gene polymorphism associates with graft-versus-host disease and survival in HLA-matched sibling allogeneic bone marrow transplantation. Bone Marrow Transplant. 2002;30:223–228. doi: 10.1038/sj.bmt.1703629. [DOI] [PubMed] [Google Scholar]

RESOURCES