KEY WORDS: Unrelated SCT, HLA-mismatch, ATG, Graft-versus-host disease

Similar documents
The impact of HLA matching on unrelated donor hematopoietic stem cell transplantation in Korean children

National Marrow Donor Program HLA-Matching Guidelines for Unrelated Marrow Transplants

KEY WORDS: Allogeneic, Hematopoietic cell transplantation, Graft-versus-host disease, Immunosuppressants, Cyclosporine, Tacrolimus

Biol Blood Marrow Transplant 17: (2011) Ó 2011 American Society for Blood and Marrow Transplantation

Does anti-thymocyte globulin have a place in busulfan/fludarabine

The National Marrow Donor Program. Graft Sources for Hematopoietic Cell Transplantation. Simon Bostic, URD Transplant Recipient

MUD SCT. Pimjai Niparuck Division of Hematology, Department of Medicine Ramathibodi Hospital, Mahidol University

Reduced-intensity Conditioning Transplantation

Cover Page. The handle holds various files of this Leiden University dissertation.

Clinical Use of Umbilical Cord Blood Hematopoietic Stem Cells

What s new in Blood and Marrow Transplant? Saar Gill, MD PhD Jan 22, 2016

Hee-Je Kim, Woo-Sung Min, Byung-Sik Cho, Ki-Seong Eom, Yoo-Jin Kim, Chang-Ki Min, Seok Lee, Seok-Goo Cho, Jong-Youl Jin, Jong-Wook Lee, Chun-Choo Kim

Haplo vs Cord vs URD Debate

Haploidentical Transplantation today: and the alternatives

Role of NMDP Repository in the Evolution of HLA Matching and Typing for Unrelated Donor HCT

Haploidentical Transplantation: The Answer to our Donor Problems? Mary M. Horowitz, MD, MS CIBMTR, Medical College of Wisconsin January 2017

HLA-DR-matched Parental Donors for Allogeneic Hematopoietic Stem Cell Transplantation in Patients with High-risk Acute Leukemia

CMV Infection after Transplant from Cord Blood Compared to Other Alternative Donors: The Importance of Donor-Negative CMV Serostatus

Trends in Hematopoietic Cell Transplantation. AAMAC Patient Education Day Oct 2014

Reduced-Intensity Conditioning Stem Cell Transplantation: Comparison of Double Umbilical Cord Blood and Unrelated Donor Grafts

Allogeneic Hematopoietic Stem Cell Transplantation: State of the Art in 2018 RICHARD W. CHILDS M.D. BETHESDA MD

Unrelated Donor Hematopoietic Cell Transplantation: Factors Associated with a Better HLA Match

Hugo Castro-Malaspina, Richard E. Harris, James Gajewski, Norma Ramsay, Robert Collins, Bernie Dharan, Roberta King, and H.

Matched and mismatched unrelated donor transplantation: is the outcome the same as for matched sibling donor transplantation?

Sylwia Mizia, 1 Dorota Dera-Joachimiak, 1 Malgorzata Polak, 1 Katarzyna Koscinska, 1 Mariola Sedzimirska, 1 and Andrzej Lange 1, 2. 1.

5/9/2018. Bone marrow failure diseases (aplastic anemia) can be cured by providing a source of new marrow

Plenary paper. Introduction

ASBMT. Impact of the Direction of HLA Mismatch on Transplantation Outcomes in Single Unrelated Cord Blood Transplantation

KEY WORDS: Comorbidity index, Reduced-intensity conditioning stem cell transplantation, Allo-RIC, HCT-CI, Mortality INTRODUCTION

Samples Available for Recipient Only. Samples Available for Recipient and Donor

Scoring HLA Class I Mismatches by HistoCheck Does Not Predict Clinical Outcome in Unrelated Hematopoietic Stem Cell Transplantation

Effect of HLA mismatch on acute graft-versus-host disease

Stem Cell Transplantation for Severe Aplastic Anemia

MATCHMAKER, MATCHMAKER, MAKE ME A MATCH, FIND ME A MISMATCHED TRANSPLANT TO CATCH

Samples Available for Recipient and Donor

Samples Available for Recipient Only. Samples Available for Recipient and Donor

HCT for Myelofibrosis

An Introduction to Bone Marrow Transplant

KEY WORDS: CRp, Platelet recovery, AML, MDS, Transplant

Outcomes From Unrelated Donor Hematopoietic Stem Cell Transplantation

Il Trapianto da donatore MUD. Alessandro Rambaldi

Allogeneic hematopoietic stem cell transplantation from family members other than HLA-identical siblings over the last decade ( )

MUD HSCT as first line Treatment in Idiopathic SAA. Dr Sujith Samarasinghe Great Ormond Street Hospital for Children, London, UK

Histocompatibility Evaluations for HSCT at JHMI. M. Sue Leffell, PhD. Professor of Medicine Laboratory Director

Hematopoietic stem cell transplantation donor sources in the 21st century: choosing the ideal donor when a perfect match does not exist

New trends in donor selection in Europe: "best match" versus haploidentical. Prof Jakob R Passweg

Introduction to Clinical Hematopoietic Cell Transplantation (HCT) George Chen, MD Thursday, May 03, 2018

Umbilical Cord Blood Transplantation

Current Status of Haploidentical Hematopoietic Stem Cell Transplantation

Myeloablative and Reduced Intensity Conditioning for HSCT Annalisa Ruggeri, MD, Hôpital Saint Antoine Eurocord- Hôpital Saint Louis, Paris

Federica Galaverna, 1 Daria Pagliara, 1 Deepa Manwani, 2 Rajni Agarwal-Hashmi, 3 Melissa Aldinger, 4 Franco Locatelli 1

The New England Journal of Medicine BONE MARROW TRANSPLANTS FROM UNRELATED DONORS FOR PATIENTS WITH CHRONIC MYELOID LEUKEMIA

T-cell-replete haploidentical HSCT with low-dose anti-t-lymphocyte globulin compared with matched sibling HSCT and unrelated HSCT

Clinical Study Steroid-Refractory Acute GVHD: Predictors and Outcomes

Back to the Future: The Resurgence of Bone Marrow??

KEY WORDS: Total body irradiation, acute myelogenous leukemia, relapse

KEY WORDS: Hematopoietic cell transplantation, Pulmonary complications

Feasibility and Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in 30 Patients with Poor Risk Acute Myeloid Leukemia Older than 60 Years

Stem Cell Transplantation

AIH, Marseille 30/09/06

Impact of HLA-Mismatch in Unrelated Donor Hematopoietic Stem Cell Transplantation: A Meta-Analysis

ATG Prevents Severe Acute Graft-versus-Host Disease in Mismatched Unrelated Donor Hematopoietic Cell Transplantation

Poor Outcome in Steroid-Refractory Graft-Versus-Host Disease With Antithymocyte Globulin Treatment

EBMT Complications and Quality of Life Working Party Educational Course

Pre-Engraftment Syndrome after Double-Unit Cord Blood Transplantation: A Distinct Syndrome not Associated with Acute Graft-Versus-Host Disease

CONSIDERATIONS IN DESIGNING ACUTE GVHD PREVENTION TRIALS: Patient Selection, Concomitant Treatments, Selecting and Assessing Endpoints

Correspondence should be addressed to Yingjun Chang;

Transplantation - Challenges for the future. Dr Gordon Cook S t James s Institute of Oncology, Leeds Teaching Hospitals Trust

Effect of Conditioning Regimen Intensity on CMV Infection in Allogeneic Hematopoietic Cell Transplantation

The question is not whether or not to deplete T-cells, but how to deplete which T-cells

Bone Marrow Transplantation and the Potential Role of Iomab-B

NIH Public Access Author Manuscript Lancet Oncol. Author manuscript; available in PMC 2011 August 29.

21/05/2018. Continuing Education. Presentation Recording. learn.immucor.com

Rob Wynn RMCH & University of Manchester, UK. HCT in Children

Disclosure. Objectives 1/22/2015

Factors Influencing Haematopoietic Progenitor cell transplant outcome Optimising donor selection

The future of HSCT. John Barrett, MD, NHBLI, NIH Bethesda MD

INTRODUCTION Rabbit antithymocyte globulin (ratg), a polyclonal antibody that targets human T lymphocytes, is

Outcomes of pediatric bone marrow transplantation for leukemia and myelodysplasia using matched. unrelated donors

Hematopoietic Cell Transplantation for Myelofibrosis. Outline

Jong Wook Lee, Byung Sik Cho, Sung Eun Lee, Ki Seong Eom, Yoo Jin Kim, Hee Je Kim, Seok Lee, Chang Ki Min, Seok Goo Cho, Woo Sung Min, Chong Won Park

Hematopoietic Engraftment in Recipients of Unrelated Donor Umbilical Cord Blood Is Affected by the CD34 and CD8 Cell Doses

Abstract. Introduction

Low-Dose Total Body Irradiation, Fludarabine, and Antithymocyte Globulin Conditioning for Nonmyeloablative Allogeneic Transplantation

journal of medicine The new england Outcomes after Transplantation of Cord Blood or Bone Marrow from Unrelated Donors in Adults with Leukemia abstract

Therapeutic Advances in Treatment of Aplastic Anemia. Seiji Kojima MD. PhD.

RIC in Allogeneic Stem Cell Transplantation

Graft-versus-host disease-free relapse-free survival, which is defined

ADVANCES IN THE MANAGEMENT OF MYELODYSPLASTIC SYNDROMES

Hematopoietic Stem Cell Transplant in Sickle Cell Disease- An update

Disclosures. Investigator-initiated study funded by Astellas

A comparison between allogeneic stem cell transplantation from unmanipulated haploidentical and unrelated donors in acute leukemia

Cord-Blood Transplantation in Patients with Minimal Residual Disease

Comparable Outcome with T-Cell Depleted Unrelated-Donor versus Related-Donor Allogeneic Bone Marrow Transplantation

Telephone: ; Fax: ; E mail:

Donor Lymphocyte Infusion for Malignancies Treated with an Allogeneic Hematopoietic Stem-Cell Transplant

Abstract. Introduction. Editor: M. Paul

Haploidentical Transplants for Lymphoma. Andrea Bacigalupo Universita Cattolica Policlinico Gemelli Roma - Italy

Outcomes of Transplantation with Related- and Unrelated-Donor Stem Cells in Children with Severe Thalassemia

a resource for physicians Recommended Referral Timing for Stem Cell Transplant Evaluation

Transcription:

HLA-Mismatched Unrelated Donors as an Alternative Graft Source for Allogeneic Stem Cell Transplantation after Antithymocyte Globulin-Containing Conditioning Regimen Nicolaus Kröger, 1 Tatjana Zabelina, 1 Thomas Binder, 2 Francis Ayuk, 1 Ulrike Bacher, 1 Gitta Amtsfeld, 1 Heinrich Lellek, 1 Johanna Schrum, 1 Rolf Erttmann, 1 Thomas Eiermann, 2 Axel Zander 1 Between August 1996 and December 2004, 369 patients with a median age of 41 years (range: 1-68 years) received stem cell transplantation (SCT) from unrelated donors after an antithymocyte-globulin (ATG)- containing conditioning regimen. In 268 patients, complete molecular typing (4-digit) of HLA-A, -B, -C, -DRB1, and -DQB1 was available: 110 patients were completely matched for 10 alleles, 91 patients had 1 allele-mismatch (9/10), and 67 patients were mismatched for 2-4 alleles (6-8/10). The incidence of grade II-IV acute graft-versus-host disease (agvhd) was 33% in the 10/10, 41% in the 9/10, and 40% in the 6-8/10 group, respectively (P 5.1). The cumulative incidence of treatment-related mortality (TRM) and relapse among the groups were similar (27%, 31%, and 32%, P 5.2; and 28%, 27%, and 26%, P 5.9. After a median follow-up of 35 months (range: 3-120 months), the estimated 5-year disease-free survival (DFS) was 42% and did not differ among the 10/10, the 9/10, and the 6-8/10-mismatched groups (45% versus 42% versus 39%) (P 5.5). In multivariate analysis, only age (hazard ratio [HR] 1.013) (P 5.004) and badrisk disease (HR 1.975) (P \.001) were independent risk factors for DFS. In conclusion, pretransplant ATG allows allogeneic SCT from unrelated donors with HLA disparities. Biol Blood Marrow Transplant 15: 454-462 (2009) Ó 2009 American Society for Blood and Marrow Transplantation KEY WORDS: Unrelated SCT, HLA-mismatch, ATG, Graft-versus-host disease INTRODUCTION Hematopoietic stem cell transplantation (SCT) from HLA-identical siblings or adult unrelated donors can cure several malignant and nonmalignant hematologic diseases and has become an established treatment. For patients lacking an HLA-identical sibling, more than 10 million unrelated volunteers worldwide are available for donation of hematopoietic stem cells. Despite this increasing number of unrelated donors, up to 30% of the patients will not find a completely From the 1 Department of Stem Cell Transplantation, University Hospital Hamburg-Eppendorf, Hamburg, Germany; and 2 Institute for Transfusion Medicine, University Hospital Hamburg- Eppendorf, Hamburg, Germany. Financial disclosure: See Acknowledgments on page 461. Correspondence and reprint requests: Nicolaus Kröger, Department of Stem Cell Transplantation, University Hospital Hamburg- Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany (e-mail: nkroeger@uke.uni-hamburg.de). Received November 23, 2008; accepted January 6, 2009 Ó 2009 American Society for Blood and Marrow Transplantation 1083-8791/09/154-0001$36.00/0 doi:10.1016/j.bbmt.2009.01.002 HLA-matching related or unrelated donor. For patients with hematologic malignancies who lack a suitable matched related or unrelated donor, encouraging results have been obtained after transplantation of hematopoietic stem cells from an HLA-haploidentical related donor [1,2], or after transplantation of 1 or 2 umbilical cord blood units either after myeloablative [3,4] or after reduced intensity conditioning (RIC) [5,6]. Both HLA-haploidentical related and umbilical cord blood grafts can be obtained rapidly for the majority of patients. For unrelated SCT, increasing genetic disparity, particularly for the human leukocyte antigen (HLA) molecular system is associated with an increased risk of acute and chronic graft-versus-host disease (agvhd, cgvhd) and a higher nonrelapse mortality (NRM), resulting in a lower survival after hematopoietic SCT [4,7-22]. Therefore, for patients without a completely HLA-matched donor, there are still questions such as how the best mismatched donor can be selected, which mismatch can be tolerated, and which GVHD prophylaxis is most appropriate. An analysis of the National Marrow Donor Program (NMDP) recently showed that allele level-matching 454

Biol Blood Marrow Transplant 15:454-462, 2009 HLA-Mismatched Donor as an Alternative Stem Cell Source 455 for HLA-A, -B, -C, and -DRB1 is preferred in the selection of adult unrelated donors, and if mismatching is unavoidable, mismatches at HLA-B or -C may be better tolerated than those in -A or -DRB1 [7]. However, in this study each single mismatch was associated with a 10% decrease in survival [7]. Therefore, transplant-related strategies such as improved graft-versushost prophylaxis, should be investigated to determine the influence of allele- and antigen-mismatched transplantation from unrelated donors. Smaller studies suggest that antithymocyte globulin (ATG) can overcome the negative effect of mismatched unrelated SCT [23-29]. In the current study, we investigated the impact of using pretransplantation ATG (ATG Fresenius Ò ) as part of the conditioning regimen on outcome of allele- and antigen-mismatched SCT from unrelated donors. PATIENTS AND METHODS Patients Between August 1996 and December 2004, 369 patients received stem cell grafts from unrelated donors at the Department of Stem Cell Transplantation of the University Hospital Hamburg-Eppendorf. In 268 cases, complete molecular typing (4-digit) of HLA-A, -B, -C, -DRB1, and -DQB1 was available for donor as well as for recipients. The median age of patients was 41 years (range: 1 68 years). Diagnoses were acute leukemia or myelodysplastic syndromes (MDS) in 135 patients, chronic myeloproliferative disease (chronic myelogenous leukemia [CML] or primary myelofibrosis [PMF]) (n 5 53), lymphoid malignancy (chronic lumphocytic leukemia [CLL], non-hodgkin leukemia [NHL], Hodgkin disease [HD], multiple myeloma [MM]) (n 5 60), or other diseases, such as aplastic anemia (AA) and nonmalignant diseases (n 5 20). Stem cell source was either bone marrow (n 5 103) or peripheral blood stem cells (PBSC; n 5 165). The patients were classified either as good risk (n 5 108; CML in first chronic phase, acute leukemia in first remission, MDS-refractory anemia [MDS-RA] or MDS-refractory anemia with ring sideroblasts [MDS-RARS], or myelofibrosis and nonmalignant diseases) or as bad risk (n 5 160; acute leukemia not in remission or more than first remission, MDS-refractory anemia with excess blasts [MDS- RAEB] or -RAEB-t, CML in accelerated phase or blastic phase, refractory NHL, and MM). The majority of the patients (n 5 163) received standard myeloablative conditioning, whereas 105 patients received a doseconditioning regimen (for details, see Table 1). The matched and the mismatched group did not differ significantly in age, sex, risk category, graft source, cytomegalovirus (CMV)-status, conditioning, or CD34 1 transplanted cells. The 6-8/10-mismatched group, however, received a significantly higher dose of ATG (ATG Fresenius Ò, Gräfelfing, Germany) (90 mg/kg body weight [BW] versus 60 mg/kg BW). According to high-resolution HLA typing, 110 of the patients were completely matched for 10 alleles, whereas 91 patients had at least 1 allele-mismatch (9/ 10), and 67 patients were mismatched for 2-4 alleles (6 8/10). Of these 67 patients, 44 had 2 mismatches, 15 had 3 mismatches, and 8 patients had 4 mismatches. The majority of the patients had mismatches in C- locus. The detailed mismatches in class I and II are listed in Table 2. HLA Typing HLA-A, -B, -C, -DRB1, and -DQB1 alleles were typed with sequence-specific oligonucleotide probes. Genomic DNA of patients and donors was isolated from 5 ml of blood using the QIAamp DNA Maxi Blood Kit (Qiagen, Chatsworth, CA). The remaining ambiguities in alleles with population frequencies.1/1000 were resolved by sequence-based typing and/or sequence specific amplication. GVHD Prophylaxis All patients received ciclosporine A (CsA) starting on day 1 at 3 mg/kg i.v. infusion and later switched to an equivalent oral dose as soon as possible. CsA was tapered off as from day 1140 and discontinued between days 1180 and 1200, if no GVHD occurred. Methotrexate (MTX; 10 mg/m 2 ) was administered on days 11, 13 and 16. agvhd and cgvhd were graded according to international standard [30,31]. Supportive Care All patients were nursed in reverse isolation in conventional or laminar air flow room. Acyclovir and fluconazol or itraconazol were routinely administed to all patients. Prophylaxis against Pneumocystis carinii consisted of cotrimoxazole or pentamidine inhalation. All blood products were irradiated with 25 Gy. CMVnegative patients received only CMV-negative blood products. CMV-positive patients were monitored at least weekly for CMV infection by polymerase chain reaction (PCR) and/or antigenemia assay. Neutropenic fever was treated with broad-spectrum antibiotics according to the centers policy. Statistical Analysis Endpoints were overall survival (OS), disease-free survival (DFS), nonrelapse mortality (NRM), incidence of relapse, incidence and severity of agvhd and cgvhd. Characteristics of patients were expressed as median and range for continous variables and frequencies for categoric variables. Categoric data were compared by chi-square test and Fisher s exact test. Actuarial curves were estimated according the Kaplan-Meier method and the significance was

456 N. Kröger et al. Biol Blood Marrow Transplant 15:454-462, 2009 Table 1. Patients Characteristics Number of Patients All Patients n 5 268 Matched 10/10 (n 5 110) Mismatched 9/10 (n 5 91) Mismatched 6-8/10 (n 5 67) P-Value Age (years) (median) 41 (range, 1-68) 41 (range, 1-67) 41 (range, 1-65) 35 (range, 1-68).2 Patients sex Male n 5 163 n 5 68 n 5 54 n 5 41.9 Female n 5 105 n 5 42 n 5 34 n 5 26 Diagnosis ALL, AML, MDS n 5 135 n 5 55 n 5 44 n 5 36.09 CML, PMF n 5 53 n 5 20 n 5 25 n 5 8 CLL, NHL, HD, MM n 5 60 n 5 29 n 5 17 n 5 14 Others n 5 20 n 5 6 n 5 5 n 5 9 Risk category Bad n 5 160 n 5 68 n 5 52 n 5 40.8 Good n 5108 n 5 42 n 5 39 n 5 27 Donors sex Male n 5 183 n 5 78 n 5 61 n 5 43.8 Female n 5 83 n 5 32 n 5 29 n 5 22 Graft source Bone marrow n 5 103 n 5 35 n 5 37 n 5 31.1 Peripheral blood n 5 165 n 5 75 n 5 54 n 5 36 stem cells AB0-compatibility Identical n 5 107 n 5 46 n 5 42 n 5 19.04 Major n 5 97 n 5 38 n 5 25 n 5 34 Minor n 5 55 n 5 24 n 5 20 n 5 11 CMV-serostatus (patients) Positive n 5 147 n 5 58 n 5 45 n 5 44.1 Negative n 5 121 n 5 52 n 5 46 n 5 23 Conditioning Standard n 5 163 n 5 62 n 5 57 n 5 44.4 Reduced intensity n 5 105 n 548 n 5 34 n 5 23 Busulfan-based Yes n 5 136 n 5 54 n 5 51 n 5 31.4 No n 5 132 n 5 56 n 5 40 n 5 36 TBI-based Yes n 5 62 n 5 24 n 5 20 n 5 18.7 No n 5 206 n 5 86 n 5 71 n 5 49 ATG-dose (median) 60 (range, 20-120) 60 (range, 20-120) 60 (range, 20-90) 90 (range, 30-90).001 CD34 + transplanted cells/kg BW (10 6 ) 6 (range, 0.35-31) 6.0 (range, 1-31) 7.0 (range, 0.35-27.0) 6.0 (range, 1-19).7 Median follow-up (months) 38 (range, 4-119) 39 (range, 4-104) 39 (range, 5-119) 32 (range, 4-97).9 ALL indicates acute lymphoblastic leukemia; AML, acute myelogenous leukemia; CLL, chronic lymphoblastic leukemia; CML, chronic myelogenous leukemia; CMV, cytomegalovirus; TBI, total body irradiation; ATG, antithymocyte globulin; BW, body weight; MM, multiple myeloma; HD, Hodgkin disease; NHL, non-hodgkin lymphoma; MDS, myelodysplastic syndromes; PMF, primary myelofibrosis. estimated by the log-rank test. OS was measured from transplantation to death from any cause. Patients who were still alive at follow-up were censored at the last follow-up date. DFS was calculated from transplantation until relapse/progression or death from any cause. Patients who were still alive at time of analysis were censored at last follow-up date. Treatment-related mortality (TRM) was determined from the date of transplantation to death related to transplantation. Patients who died from other causes were censored at time of death. Estimates for relapse incidence and TRM were made using cumulative incidence estimates. The following factors were included: age, HLA-match, diagnosis, standard conditioning versus RIC, good versus bad risk, total body irradiation (TBI) versus non-tbi, graft source. Factors that had value of P \.05 were entered in a multivariable Cox regression model (forward elimination) to determine independent predictors. The results were presented as hazard ratio (HR) together with the 95% confidence interval (CI) and P-values. Calculations were performed in SPSS version 12.0 (SPSS, Chicago, IL). The cumulative incidences were analyzed with the Gray-test (according to National Health and Medical Research Council, Clinical Trial Center, University of Sydney, 2007, Prof. Val Gebski). RESULTS Engraftment All but 2 patients experienced stable engraftment of leukocytes and platelets. Two patients (1%) who experienced primary graft failure had been transplanted from 10/10-matched donors, whereas none of the mismatch-transplanted patients had primary graft failure. The median time to leukocyte engraftment was 16 days in the matched as well as in the mismatched group (range: 11 29 days, and 10-27 days, respectively) (P 5.9). The median time to platelet engraftment (.20 10 9 /L) was 23 days (range: 9-145 days) for

Biol Blood Marrow Transplant 15:454-462, 2009 HLA-Mismatched Donor as an Alternative Stem Cell Source 457 Table 2. Characteristics of HLA-Mismatches (mm) Class I mm n 5 134 Class II mm n 5 54 1 mm class I/II n 5 91 2 mm class I/II n 5 44 3 mm class I/II n 5 15 4 mm class I/II n 5 8 Class I A-mismatch n 5 17 Allele mismatch n 5 12 Antigen mismatch n 5 5 B-mismatch n 5 40 Allele mismatch n 5 31 Antigen mismatch n 5 9 C-mismatch n 5 116 Allele mismatch n 5 14 Antigen mismatch n 5 95 Allele and antigen mismatch n 5 7 KIR-ligand mismatch n 5 39 Overall class I 1 mismatch n 5 87 >2 mismatches n 5 47 Class II DR-mismatch n 5 29 Allele mismatch n 5 24 Antigen mismatch n 5 5 DQ-mismatch n 5 35 Allele mismatch n 5 13 Antigen mismatch n 5 22 Overall class II 1 mismatch n 5 44 $2 mismatches n 5 10 the 10/10-matched group, 21 days (range: 11-90 days) for the 9/10-mismatched group, and 18 days (range: 11-132 days) for the 6 8/10 mismatched group (P 5 0.8). GVHD Overall, 42% of the study population did not experience any agvhd. No agvhd was seen in 49% the 10/10-matched group, in 40% of the 9/10-mismatched group, and in 35% of the 6 8/10-mismatched group. The incidence of grade II-IV agvhd was 33% of the 10/10-matched group, 41% in the 9/10-mismatched-group, and 40% in the 6-8/10-mismatchedgroup (P 5.1). Severe grade III-IV agvhd was seen in 18% of the 10/10-matched group, in 15% of the 9/ 10-mismatched-group, and in 23% of the 6-8/10-mismatched group. The overall rate of cgvhd was 42% in the 10/10-matched group, 37% in the 9/10-mismatched group, and 46% in the 6-8/10-mismatched group (P 5.8). Severe extensive GVHD was seen in 17% of the 10/10-matched patients, and in 19% each in the 9/10-mismatched and in the 6-8/10-mismatched groups (P 5.8) (Table 3). TRM The cumulative incidence of TRM at 3 years for the whole study group was 30% (95% CI: 25-35). Causes of death were infectious complications (45%), toxicity (25%), GVHD (25%), posttransplant lymphoma (3%), graft failure (1%), and secondary cancer (1%). TRM in the 10/10-matched group was 27%; in the 9/10-mismatched group 31%, and in the 6-8/10- mismatched group 33% (P 5.2) (Figure 1). Significant factors for higher TRM in the univariate analysis were age, either as continuous variables or, in case cutoffs were used, as variables. Patients with good-risk disease had a significantly lower treatment-related mortality compared to patients with bad-risk disease (24% versus 39%) (P 5.02) (Table 4), whereas HLA-matched versus HLA-mismatched did not influence TRM (29% versus 34%) (P 5.2). Even patients with bad risk disease, the treatment-related mortality did not differ between the 10/10-matched, the 9/10-mismatched, and the 6-8/10-mismatched group (34% versus 38% versus 51%) (P 5.4). In a multivariate analysis, only age as a continuous variable (HR 1.023) (P \.001) significantly influenced TRM (Table 5). Relapse The cumulative incidence of relapse at 5 years for the entire study population was 27% (95% CI: 21-33%). There was no difference in the cumulative incidence of relapse between the 10/10-matched, the 9/10-mismatched, and the 6-8/10-mismatched group (28% versus 27% versus 26%) (P 5.9) (Figure 2). There was also no significant difference in the cumulative incidence of relapse if mismatching was restricted only to patients with bad risk (49% versus 53% versus 47%) (P 5 0.3). In an univariate analysis, significant factors for higher relapse incidence (cumulative incidence at 5 years) were lymphoid malignancies: NHL, MM, CLL, HD (70%) versus acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), MDS (36%) versus CML, myelofibrosis (24%) (P 5.01). Other risk factors for higher incidence of relapse within the univariate analysis were bad-risk disease in comparison to goodrisk disease (51% versus 18%) (P\.001), and RIC versus standard intensity conditioning (62% versus 26%) (P 5.001). In the multivariate analysis, the only significant factors for a higher incidence of relapse at 5 years were bad-risk diseases (HR 5 2.966) (P \.001). DFS After a median follow-up of 35 months (range: 3-120 months), the 5-year estimated DFS rate for the entire study group was 42%. It did not differ among the 10/10-matched, the 9/10-mismatched, and the 6-8/10-mismatched groups (45% versus 42% versus 39%) (P 5.5) (Figures 3 and 4). In the univariate analysis, significant factors for an improved DFS were younger age (P 5.001), nonmalignant disease (P 5.002), good-risk disease (P \.001), and standard conditioning (P 5.006). In the multivariate analysis, only age (HR 5 1.013) (P 5.004) and bad-risk disease (HR 5 1975) (P \.001) remained independent risk factors for DFS.

458 N. Kröger et al. Biol Blood Marrow Transplant 15:454-462, 2009 Table 3. Results All Patients n 5 268 Matched 10/10 (n 5 110) Mismatched 9/10 (n 5 91) Mismatched 6-8/10 (n 5 67) P-Value Primary graft-failure n 5 2 (1%) n 5 2 n 5 0 n 5 0.2 Median leukocyte 16 days (range, 10-38) 16 days (range, 10-38) 16 days (range, 11-29) 16 days (range, 10-27).9 engraftment (>1.0 10 9 / L) Median platelet 21 days (range, 9-145) 23 days (range, 9-145) 21 days (range, 11-90) 18 days (range, 11-132).8 engraftment (>20 10 9 / L) (median) Acute graft-versus-host.1 disease (agvhd) Grade 0 n 5 112 (42%) n 5 53 (49%) n 5 36 (40%) n 5 23 (35%) Grade 1 n 5 54 (20%) n 5 20 (19%) n 5 18 (20%) n 516 (25%) Grade 2 n 5 50 (18%) n 5 16 (15%) n 5 23 (26%) n 5 11 (17%) Grade 3 n 5 31 (12%) n 5 16 (15%) n 5 6 (7%) n 5 8 (12%) Grade 4 n 5 17 (6%) n 5 3 (3%) n 5 7 (8%) n 5 7 (11%) Chronic graft-versus-host.8 disease (cgvhd) None n 5 123 (58%) n 5 53 (58%) n 5 37 (55%) n 5 33 (63%) Limited n 5 50 (24%) n 5 23 (25%) n 5 18 (27%) n 5 9 (17%) Extensive n 5 40 (19%) n 5 16 (17%) n 5 13 (19%) n 5 10 (19%) Cumulative incidence 30% (95% CI: 25-35%) 27% (95% CI: 19-35%) 31% (95% CI: 21-41%) 33% (95% CI: 21-45%).2 of TRM (3 years) Cumulative incidence 27% (95% CI: 21-33%) 28% (95% CI: 18-38%) 27% (95% CI: 17-37%) 26% (95% CI: 14-38%).9 of relapse (5 years) Estimated disease-free 42% (95% CI: 36-48) 45% (95% CI: 35-55%) 42% (95% CI: 31-53%) 39% (95% CI: 26-52%).5 survival (DFS) (5 years) Estimated overall 51% (95% CI: 45-57) 53% (95% CI: 43-63%) 55% (95% CI: 44-66%) 41% (95% CI: 28-54%).3 survival (OS) (5 years) Estimated OS For good-risk patients 67% (95% CI: 58-76) 67% (95% CI: 53-81%) 72% (95% CI: 58-86%) 62% (95% CI: 40-84%).9 For bad-risk patients 40% (95% CI: 32-48) 45% (95% CI: 29-61%) 43% (95% CI: 29-57%) 26% (95% CI: 10-42%).2 Estimated DFS For good-risk patients 61% (95% CI: 51-71) 62% (95% CI: 47-77%) 64% (95% CI: 49-79%) 59% (95% CI: 37-81%).9 For bad-risk patients 29% (95% CI: 21-37) 33% (95% CI: 19-47%) 28% (95% CI: 14-42%) 26% (95% CI: 12-40%).2 Estimated incidence of relapse For good-risk patients 18% (95% CI: 9-27) 20% (95% CI: 6-34%) 16% (95% CI: 2-30%) 18% (95% CI: 0-38%).7 For bad-risk patients 51% (95% CI: 40-62) 49% (95% CI: 31-67%) 53% (95% CI: 37-73%) 47% (95% CI: 27-67%).3 Estimated incidence of TRM For good-risk patients 24% (95% CI: 16-32) 23% (95% CI: 9-39%) 24% (95% CI: 10-38%) 22% (95% CI: 6-38%).9 For bad-risk patients 39% (95% CI: 31-47) 34% (95% CI: 21-47) 38% (95% CI: 24-52%) 51% (95% CI: 31-71%).4 TRM indicates treatment-related mortality. OS The estimated 5-year OS rate was 41% (95% CI: 45%-57%). There was no statistical difference among the 10/10-matched, the 9/10-mismatched, and the 6-8/10-mismatched groups (53% versus 55% versus 41%) (P 5.3) (Figures 3 and 4). In the univariate analysis, significant factors for an improved OS were younger age (P \.001), nonmalignant disease (P \.001), good-risk disease (P \.001), and standard-intensity conditioning (P 5.03). In a multivariate analysis, age as continuous variable (HR 5 1.017) (P 5.001), and bad-risk disease (HR 5 1.920) (P 5.001) remained independent significant factors for OS. Influence of HLA-Mismatch We analyzed locus or class mismatches and mismatch combinations with respect to treatment-related mortality, relapse, and survival. For the majority of single mismatches the number of patients was too small to perform valid analysis. The only significant factor was a combined B- and A-allele-mismatches resulted in a significantly worse OS in comparison to 10/10-matched patients (35% versus 53%) (P 5.04). All other single mismatches or mismatch combinations did not influence treatment-related mortality, relapse, DFS, and OS. DISCUSSION This large single-center experience using ATG as part of the conditioning regimen for unrelated SCT showed that up to 4 mismatches either on the alleleor antigen-level can be tolerated without an obvious increased risk of agvhd and cgvhd or TRM, resulting in a comparable OS and DFS as being achieved for patients with high-resolution completely matched- HLA-A, -B, -C, -DRB1, and -DQB1-matched hematopoietic stem cell donors. This is 1 of the first reports suggesting that by modifying the GVHD-prophylaxis,

Biol Blood Marrow Transplant 15:454-462, 2009 HLA-Mismatched Donor as an Alternative Stem Cell Source 459 Figure 1. Cumulative incidence of TRM according to HLA-matching. mismatched unrelated stem cell donor grafts can be tolerated without worsening survival rates. Similar results have been reported previously, but the respective analyses were based only on serologic class I-typing (without HLA C) and high-resolution typing only for class II. These results are of interest because there is general agreement that increasing genetic disparity on HLA-molecules is associated with a higher risk of agvhd, a higher TRM, and a lower survival in unrelated SCT [7,8,10-12,14-18,20-22]. However, which antigen- or allele-mismatches are of most clinical relevance is still a matter of debate. A recent analysis of the NMDP, which included 3857 transplant procedures, suggested that high-resolution DNA-matching for HLA-A, -B, -C, or -DRB1 (8/8) was confirmed as minimum matching-level associated with highest survival. A single mismatch detected either by low- or highresolution DNA testing at HLA-A, -B, -C, or -DRB1 (7/8) was associated with a higher mortality, and a 1-year survival rate of 43% compared to 55% for the 8/8-matched pairs. In this analysis, each mismatch was associated with a reduced survival, but mismatches at HLA-B or -C may be better tolerated than those on HLA-A or -DRB1. Furthermore, there was no difference whether the mismatch was seen at the allele-level or at the antigen-level, except for HLA-C, in which the allele-mismatch was better tolerated [7]. This analysis was restricted only to standard-myeloablative conditioning for patients with AML, ALL, CML, and MDS, and the stem cell source was primarily bone marrow. In contrast to our study, in which all patients received ATG, in the NMDP study, only 22% of the patients received some form of T cell-depleted grafts, but no detailed information about in vivo or in vitro T cell depletion was given. In another study from the Fred Hutchinson Cancer Center in Seattle, HLA- DRB1 allele-mismatch was associated with a high rate of agvhd, and of mortality [11]. Later on, the same group reported on 471 CML patients who underwent allogeneic SCT from an unrelated donor, and they reported that single-antigen, but not alleleclass I mismatches, were associated with an increased risk of graft failure, whereas the survival at 5 years did not differ significantly among single class I allele- Table 4. Univariate Analysis of Overall Survival (OS), Disease-Free Survival (DFS), Relapse, and Treatment-Related Mortality at 5 Years (269 Patients) OS at 5 Years % (Range) P- value DFS at 5 Years % (Range) P- Value Relapse at 5 Years % (Range) P- Value Treatment-Related Mortality at 3 Years % (Range) P-Value Age 0-20 years 75 (64-86) 63% (51-75) 14 (6-22) 21-50 years 42 (33-51) <.001 36% (27-45).001 36 (27-45).003 51-68 years 42 (30-54) 31% (18-44) 43 (30-56) Diagnosis ALL, AML, 44 (35-53) 42 (33-51) 36 (26-46) MDS 70 (58-82) 58 (44-72) 24 (10-38) CML, PMF 35 (21-49) <.001 16 (4-28).002 70 (50-90).01 NHL, MM, CLL, HD NMD 86 (71-100) 70 (50-90) 11 (0-25) Risk factor Good risk 67 (58-76) <.001 61 (51-71) <.001 18 (10-26) <.001 24 (16-32).02 Bad risk 40 (32-48) 29 (21-37) 51 (40-62) 39 (31-47) Conditioning Standard intensity 57 (49-65) 51 (43-59) 26 (18-34) Reduced intensity 38 (27-49).03 23 (12-34).006 62 (45-79).001 Age (contin.) HR 1.020 <.001 HR 1.017 <.001 HR 1.023 <.001 (95% CI: 1.010-1.030) (95% CI: 1.008-1.026) (95% CI: 1.010-1.036) HLA-mismatch 10/10 match 53 (43-63).2 45 (33-57).2 36 (24-48).7 29 (20-38).2 Mismatch 49 (41-57) 41 (33-49) 37 (27-47) 34 (26-42) ALL indicates acute lymphoblastic leukemia; AML, acute myelogenous leukemia; CLL, chronic lymphoblastic leukemia; CML, chronic myelogenous leukemia; CMV, cytomegalovirus; MM, multiple myeloma; HD, Hodgkin disease; NHL, non-hodgkin lymphoma; MDS, myelodysplastic syndromes; PMF, primary myelofibrosis; NMD, Non-malignant diseases.

460 N. Kröger et al. Biol Blood Marrow Transplant 15:454-462, 2009 Table 5. Multivariate Analysis of Treatment-Related Mortality, Relapse, Disease-Free Survival (DFS), and Overall Survival (OS) at 5 Years (269 Patients) Treatment- Related Mortality at 3 Years HR (95% CI) P-Value Relapse at 5 Years HR (95% CI) P-Value DFS at 5 Years HR (95% CI) P-Value OS at 5 Years HR (95% CI) P-Value Age (contin.) 1.023 (1.010-1.036) <.001 1.013 (1.004-1.022) Bad-risk disease 2.966 <.001 1.975 (1.682-5.229) (1.004-1.022).003 1.017 (1.007-1.027) <.001 1.920 (1.283-2.859).001.001 mismatched pairs, single class I antigen-mismatched pairs, and matched patients [10]. In 2004, the same group extended their analysis to 948 patients and reported single allele- or antigen-mismatch was associated with a higher mortality in CML in chronic phase within 2 years of diagnosis, but not in patients with more advanced disease. In detail, a single disparity at HLA-C was associated with a lower survival than it was with HLA-DQB1-mismatch in combination with other mismatches. As in the recently published NMDP study, the Seattle group also found that a single allele and a single antigen mismatch had similar mortality results. In contrast to this group, the NMDP study has also clearly shown a lower survival for mismatched transplantation in patients with advanced disease phases. Another large study from the Japanese Marrow Donor Program ( JMDP) reported on 1298 SCTs. It found that each mismatch on HLA-A, -B, -C, or -DRB1 and -DQB1 was associated with a higher risk of grade III-IV agvhd, but that only HLA-A and -B mismatches were associated with a higher mortality, and with cgvhd [8]. Besides the aforementioned NMDP study from 2007, the NMDP published 2 earlier studies examining the HLA-matching in adult unrelated donor hematopoietic SCT. In 1 analysis, 831 patients with CML in chronic or accelerated phase were investigated, and HLA-DRB1 allele mismatching was associated with a lower relapse-free survival (RFS) as well as with lower OS [17]. Later on, the NMDP published another study analyzing 1874 transplantations, and reported that low-resolution mismatches at HLA- A, -B, -C, and -DRB1 were associated with a worse survival, whereas mismatches detected with highresolution typing were not associated with a lower survival [19]. Other studies including less than 200 patients confirmed the negative impact of HLA-mismatching on A probability of disease-free survival 1,0 0,8 0,6 0,4 0,2 0,0 =< 8/10 match 9/10 match 10/10 match 0 1000 2000 3000 days after transplantation 4000 B 1,0 probability of overall survival 0,8 0,6 0,4 0,2 =< 8/10 match 10/10 match 9/10 match 0,0 0 1000 2000 3000 days after transplantation 4000 Figure 2. Cumulative incidence of relapse according to HLA-matching. Figure 3. DFS (A) and OS (B) of 10/10-allele-matched, 9/10-allelemismatched, and 6-8/10-allele-mismatched unrelated donor SCT.

Biol Blood Marrow Transplant 15:454-462, 2009 HLA-Mismatched Donor as an Alternative Stem Cell Source 461 Both the incidence of agvhd and cgvhd, as well as graft failure, influenced mostly the negative outcome of mismatched transplantation in the aforementioned studies. In contrast to other T cell-depleting methods [17], there was no obvious increase of relapse using ATG as GVHD prophylaxis, but because no non-atg control group existed, no valid information can be given for ATG and an increased risk of relapse. A Chinese group that transplanted mainly leukemia patients with multiple HLA-mismatched related donors without in vitro T cell depletion reported a similar observation. Besides the combined bone marrow and peripheral blood SCT, the group also used high-dose ATG to prevent agvhd, and it found no influence of the occurrence of GVHD by the HLA-disparity [29]. We conclude that the current study provides evidence that the negative impact of HLA-disparity in allogeneic SCT from unrelated donor can be overcome by using ATG to prevent GVHD and primary graft failure. This treatment strategy offers new transplant possibilities for patients lacking an HLA-identical sibling or a fully matched unrelated donor. ACKNOWLEDGMENTS Figure 4. DFS (A) and OS (B) in good-risk or bad-risk disease according to HLA-matching. clinical outcome after transplantation from unrelated donors [20-22]. Therefore, almost all studies published so far confirmed the negative influence of HLA-mismatch on outcome after allogeneic SCT from unrelated donors. One might ask why our study did not show similar decrements in survival depending on HLA-mismatch. Similar to other studies, the influence of unrelated SCT in a multivariate analysis was mainly influenced by age and disease status, indicating that no selection bias of the patients could be the reason. In contrast to all other reported studies, all our patients received ATG as part of the conditioning regimen to prevent agvhd. A positive effect of ATG to prevent GVHD has been reported in several studies [23-28]. In some of those studies, small portions of mismatched-unrelated donors were included with a similar outcome to those with completely HLA-matched donor [27,28]. Therefore, the main reason for the encouraging results in mismatched-unrelated SCT is probably because of the similar incidence of agvhd and cgvhd in the 10/10-matched and the mismatched cohort. Further more, it is of note that no graft failure was observed after HLA-mismatched transplantation. We thank the staff of the BMT unit for providing excellent care to our patients, the medical technicians for their excellent work in the laboratories, and Karen Klemt for proofreading Authorship N.K. designed the study, performed analysis, and wrote the manuscript. T.Z. performed statistical analysis, T.B. and T.E. performed HLA typing, F.A., U.B., G.A., H.L., J.S., and H.K. provided patient data and discussed the results. A.Z. interpreted the results. All authors approved the manuscript. Financial disclosure: The authors have nothing to disclose. REFERENCES 1. Aversa F, Terenzi A, Tabilio A, et al. Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse. J Clin Oncol. 2005;20(23):3447-3454. 2. Lu DP, Dong L, Wu T, et al. Conditioning including antithymocyte globulin followed by unmanipulated HLA-mismatched/ haploidentical blood and /marrow transplantation can achieve comparable outcomes with HLA-identical sibling transplantation. Blood. 2006;107:3065-3073. 3. Barker JN, Davies SM, DeFor T, et al. Survival after transplantation of unrelated donor umbilical cord blood is comparable to that of human leukocyte antigen-matched unrelated donor bone marrow: results of a matched-pair analysis. Blood. 2001;97: 2957-2961. 4. Laughlin MJ, Barker J, Bambach B, et al. Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors. N Engl J Med. 2001;344: 1815-1822.

462 N. Kröger et al. Biol Blood Marrow Transplant 15:454-462, 2009 5. Barker JN, Weisdorf DJ, DeFor TE, et al. Rapid and complete donor chimerism in adult recipients of unrelated donor umbilical cord blood transplantation after reduced-intensity conditioning. Blood. 2003;102:1915-1919. 6. Ballen KK, Spitzer TR, Yeap BY, et al. Double unrelated reduced-intensity umbilical cord blood transplantation in adults. Biol Blood Marrow Transplant. 2007;13:82-89. 7. Lee SJ, Klein J, Haagenson M, et al. High-resolution donorrecipient HLA matching contributes to the success of unrelated donor marrow transplantation. Blood. 2007;110:4576-4583. 8. Morishima Y, Sasazuki T, Inoko H, et al. The clinical significance of human leukocyte antigen (HLA) allele compatibility in patients receiving a marrow transplant from serologically HLA-A, HLA-B, and HLA-DR matched unrelated donors. Blood. 2002;99:4200-4206. 9. Petersdorf EW, Anasetti C, Martin PJ, et al. Limits of HLA mismatching in unrelated hematopoietic cell transplantation. Blood. 2004;104:2976-2980. 10. Petersdorf EW, Hansen JA, Martin PJ, et al. Major-histocompatibility-complex class I alleles and antigens in hematopoietic-cell transplantation. N Engl J Med. 2001;345:1794-1800. 11. Petersdorf EW, Longton GM, Anasetti C, et al. The significance of HLA-DRB1 matching on clinical outcome after HLA-A, B, DR identical unrelated donor marrow transplantation. Blood. 1995;86:1606-1613. 12. Petersdorf EW, Smith AG, Mickelson EM, et al. The role of HLA-DPB1 disparity in the development of acute graft-versus-host disease following unrelated donor marrow transplantation. Blood. 1993;81:1923-1932. 13. Rocha V, Labopin M, Sanz G, et al. Acute Leukemia Working Party of European Blood and Marrow Transplant Group; Eurocord-Netcord Registry. Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia. N Engl J Med. 2004;351:2276-2285. 14. Drobyski WR, Klein J, Flomenberg N, et al. Superior survival associated with transplantation of matched unrelated versus one-antigen-mismatched unrelated or highly human leukocyte antigen-disparate haploidentical family donor marrow grafts for the treatment of hematologic malignancies: establishing a treatment algorithm for recipients of alternative donor grafts. Blood. 2002;99:806-814. 15. Sasazuki T, Juji T, Morishima Y, et al. Effect of matching of class I HLA alleles on clinical outcome after transplantation of hematopoietic stem cells from an unrelated donor. Japan Marrow Donor Program. N Engl J Med. 1998;339:1177-1185. 16. Shaw BE, Marsh SG, Mayor NP, et al. HLA-DPB1 matching status has significant implications for recipients of unrelated donor stem cell transplants. Blood. 2006;107:1220-1226. 17. Gajewski J, Gjertson D, Cecka M, et al. The impact of T-cell depletion on the effects of HLA DR beta 1 and DQ beta allele matching in HLA serologically identical unrelated donor bone marrow transplantation. Biol Blood Marrow Transplant. 1997;3:76-82. 18. Petersdorf EW, Kollman C, Hurley CK, et al. Effect of HLA class II gene disparity on clinical outcome in unrelated donor hematopoietic cell transplantation for chronic myeloid leukemia: the US National Marrow Donor Program Experience. Blood. 2001;98:2922-2929. 19. Flomenberg N, Baxter-Lowe LA, Confer D, et al. Impact of HLA class I and class II high-resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplantation outcome. Blood. 2004;104:1923-1930. 20. Tiercy JM, Passweg J, van Biezen A, et al. Chronic Leukemia Working Party of the European Blood and Marrow Transplant Group (EBMT). Isolated HLA-C mismatches in unrelated donor transplantation for CML. Bone Marrow Transplant. 2004; 34:249-255. 21. Greinix HT, Fae I, Schneider B, et al. Impact of HLA class I high-resolution mismatches on chronic graft-versus-host disease and survival of patients given hematopoietic stem cell grafts from unrelated donors. Bone Marrow Transplant. 2005; 35:57-62. 22. Ho VT, Kim HT, Liney D, et al. HLA-C mismatch is associated with inferior survival after unrelated donor non-myeloablative hematopoietic stem cell transplantation. Bone Marrow Transplant. 2006;37:845-850. 23. Bacigalupo A, Lamparelli T, Bruzzi P, et al. Antithymocyte globulin for graft-versus-host disease prophylaxis in transplants from unrelated donors: 2 randomized studies from Gruppo Italiano Trapianti Midollo Osseo (GITMO). Blood. 2001;98: 2942-2947. 24. Zander AR, Kröger N, Schleuning M, et al. ATG as part of the conditioning regimen reduces transplant-related mortality (TRM) and improves overall survival after unrelated stem cell transplantation in patients with chronic myelogenous leukemia (CML). Bone Marrow Transplant. 2003;32:355-361. 25. Zander AR, Zabelina T, Kröger N, et al. Use of a five-agent GVHD prevention regimen in recipients of unrelated donor marrow. Bone Marrow Transplant. 1999;23:889-893. 26. Kröger N, Zabelina T, Krüger W, et al. Anti-thymocyte-globulin as part of the preparative regimen prevents graft failure and severe graft versus host disease (GvHD) in allogeneic stem cell transplantation from unrelated donors. Ann Hematol. 2001;80: 209-215. 27. Finke J, Schmoor C, Lang H, et al. Matched and mismatched allogeneic stem-cell transplantation from unrelated donors using combined graft-versus-host disease prophylaxis including rabbit anti-t lymphocyte globulin. J Clin Oncol. 2003;21: 506-513. 28. Ayuk F, Diyachenko G, Zabelina T, et al. Anti-thymocyte globulin overcomes the negative impact of HLA mismatching in transplantation from unrelated donors. Exp Hematol. 2008 May 2 [Epub ahead of print]. 29. Huang XJ, Liu DH, Liu KY, et al. Haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion for the treatment of hematological malignancies. Bone Marrow Transplant. 2006;38:291-297. 30. 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. 31. Shulman HM, Sullivan KM, Weiden PL, et al. Chronic graftversus-host syndrome in man. A long-term clinicopathologic study of 20 Seattle patients. Am J Med. 1980;69:204-217.