Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Dec 1.
Published in final edited form as: Bone Marrow Transplant. 2016 Aug 15;51(12):1599–1601. doi: 10.1038/bmt.2016.215

Comparable outcomes with marrow or peripheral blood as stem cell sources for hematopoietic cell transplantation from haploidentical donors after non-ablative conditioning: A matched-pair analysis

Paul V O'Donnell 1,2,*, Mary Eapen 3, Mary M Horowitz 3, Brent R Logan 3, Alyssa DiGilio 3, Claudio Brunstein 4, Ephraim J Fuchs 5, Mary ED Flowers 1, Rachel Salit 1, Kavita Raj 6,7, Antonio Pagliuca 7, Kenneth Bradstock 8, Angela Granata 9, Luca Castagna 10, Sabine Furst 9, Didier Blaise 9
PMCID: PMC5143194  NIHMSID: NIHMS826294  PMID: 27526284

In the setting of myeloablative conditioning and conventional post-grafting immunoprophylaxis for graft-versus-host disease (GvHD) with a calcineurin inhibitor and methotrexate, randomized studies of bone marrow (BM) versus peripheral blood (PB) as the allograft source in transplantation from HLA-matched related donors (1) or unrelated donors (2) showed comparable outcomes with the exception of an absolute increase of 6-15% in the incidence of chronic GvHD after transplantation of PB. However, in the setting of non-ablative conditioning, a recent retrospective study from the Center for International Blood and Marrow Transplant Research (CIBMTR) showed that rates of acute GvHD, chronic GvHD and overall survival were similar after transplantation of BM compared to PB (3). In order to test whether outcomes were similar after haplo-BM and haplo-PB transplants using non-ablative conditioning with the Hopkins regimen (4), we compared data from a multi-center phase II trial of haplo-BM conducted in the US (Blood and Marrow Transplant Clinical Trials Network 0603 [5,6]) to published and unpublished data from phase II trials of haplo-PB transplants in the US, Europe and Australia (7-9) by means of a matched-pair analysis. Patients 70 years of age or younger who met the eligibility criteria for BMT CTN 0603 were matched for age ± 10 years and for low-intermediate-high disease risk index (DRI). DRI is a composite of disease, disease status and cytogenetic risk (acute leukemia and MDS) that has been shown to independently risk stratify survival in heterogeneous adult patient cohorts with hematologic malignancy regardless of conditioning intensity or graft source (10).

Patient, disease and transplant characteristics of the cohorts are shown in Table 1. The median age at transplantation was 49 years. Recipients of haplo-PB transplants were more likely to report 3 or more significant co-morbidities using the HCT-CI score compared to haplo-BM transplants (47% vs. 28%). As shown in Table 1 there were differences in distribution of diseases transplanted and disease status at transplantation. However, approximately 75% of patients in both cohorts were assigned intermediate DRI. Follow-up was substantially longer in the haplo-BM compared to haplo-PB cohort (median 60 months versus 39 months, respectively). Of the 19 patients alive at last follow-up after haplo-BM transplant, 17 (89%) have over 3 years of follow-up and the remaining 2 patients, 30 and 32 months; among haplo-PB recipients, 18 of 29 patients (62%) have over 3 years of follow up, 6 patients (21%) and 5 patients (17%) have been followed for 2-3 years and 1-2 years, respectively.

Table 1. Patient and disease characteristics.

Characteristics Haplo-BMa,c Haplo-PBb,c
Number 43 43
Median age, years 49 (7 – 70) 49 (14 – 68)
Age, years
 ≤ 18 3 (7%) 2 (5%)
 19 – 40 12 (25%) 13 (30%)
 41 – 60 19 (45%) 17 (39%)
 61 – 70 10 (23%) 11 (26%)
Sex
 Male 29 (67%) 25 (58%)
 Female 14 (33%) 18 (42%)
Cytomegalovirus serostatus
 Positive 12 (28%) 12 (28%)
 Negative 31 (72%) 31 (72%)
Diseases
 Acute myeloid leukemia 20 (47%) 13 (30%)
 Acute lymphocytic leukemia 4 (9%) 3 (7%)
 Undifferenciated leukemia 3 (7%)
 Diffuse large B-cell lymphoma 6 (14%) 6 (14%)
 Mantle cell lymphoma 2 (5%) 1 (2%)
 Anaplastic large cell lymphoma __ 5 (11%)
 Peripheral T cell lymphoma __ 3 (7%)
 Mycosis fungoides __ 1 (2%)
 Non Hodgkin lymphoma, unclassifed __ 3 (7%)
 Hodgkin lymphoma 7 (16%) 8 (19%)
Disease status
 Complete remission 33 (77%) 34 (79%)
 Partial remission 10 (23%) 9 (21%)
Disease risk index
 Low risk 5 (12%) 5 (12%)
 Intermediate risk 32 (74%) 32 (74%)
 High risk 6 (14%) 6 (14%)
Co-morbidity index
 0 – 2 31 (72%) 23 (54%)
 ≥ 3 12 (28%) 20 (47%)
Prior autologus transplantation 9 (21%) 14 (33%)
a

Data obtained from the Data Coordinating Center for the BMT CTN and CIBMTR. Transplantation occurred between December 2008 and May 2010.

b

Data obtained from the combined databases of the Institut Paoli Calmettes, Marseille (22 patients), Fred Hutchinson Cancer Research Center, Seattle (9 patients), Westmead Hosptial, Sydney (7 patients) and Guy's and St. Thomas' Hospital/King's College Hospital, London (5 patients). Transplantation occurred between 2009 and 2014.

c

Institutional Review Boards of each participating institution and the National Donor Program approved the sharing of de-identified patient data in this study

As reported previously (5, 7, 8), the median time to neutrophil recovery was 17 days (range: 12 – 83 days) after haplo-BM and 18 days (range: 13-30 days) after haplo-PB transplants; the median time to platelet recovery was 25 days (range: 8-92 days) after haplo-BM and 24 days (range: 12-134 days) after haplo-PB transplants. Donor chimerism (≥95% at D28) was 86% after haplo-BM and 93%, after haplo-PB transplants.

Table 2 shows transplantation outcomes by treatment group. There were no significant differences in the rates of grade II-IV acute GvHD after haplo-BM and haplo-PB transplants. None of the recipients of haplo-BM transplants developed grade III-IV acute GVHD. The day-100 incidence of grade III-IV acute GvHD after haplo-PB transplantation was 5% (95% CI 1-14). Similarly, the 2-year incidence of chronic GvHD or its severity did not differ after transplantation of haplo-BM and haplo-PB. Among recipients of haplo-BM, global severity (11) was graded as mild (n=1), moderate (n=7) and severe (n=2). Global severity was graded as mild (n=4), moderate (n=2), and severe (n=1) among recipients of haplo-PB. No new cases of chronic GvHD were observed after 2 year post-transplant in either cohort. There was no significant difference in the incidence of non-relapse mortality and overall survival but the incidence of relapse was lower after haplo-PB compared to haplo-BM transplants.

Table 2. Univariate analyses for matched pairs: haplo-BM vs. haplo-PBa.

Outcomes Haplo-BM Haplo-PB p-value
Number 43 43
Acute grade II-IV GvHD @ day-100 33% (95% CI 19-47) 40% (95% CI 26-54) 0.50
Chronic GvHDb
 @ 1-year 21% (95% CI 10-34) 14% (95% CI 5-26) 0.39
 @ 2-year 23% (95% CI 12-37) 19% (95% CI 9-32) 0.63
 @ 3-year 23% (95% CI 12-37) 19% (95% CI 9-32) 0.63
Non-relapse mortality
 @ 1-year 5% (95% CI 0-13) 9% (95% CI 3-20) 0.40
 @ 2-year 7% (95% CI 1-16) 12% (95% CI 4-23) 0.45
 @ 3-year 7% (95% CI 1-16) 12% (95% CI 4-23) 0.45
Relapse
 @ 1-year 49% (95% CI 34-64) 19% (95% CI 9-31) 0.002
 @ 2-year 51% (95% CI 36-66) 24% (95% CI 12-38) 0.006
 @ 3-year 58% (95% CI 43-73) 24% (95% CI 12-38) <0.001
Disease-free survival
 @ 1-year 47% (95% CI 32-61) 72% (95% CI 58-84) 0.01
 @ 2-year 42% (95% CI 28-57) 65% (95% CI 50-78) 0.03
 @ 3-year 35% (95% CI 21-49) 65% (95% CI 50-78) 0.004
Overall survival
 @ 1-year 72% (95% CI 58-84) 81% (95% CI 66-90) 0.30
 @ 2-year 58% 66% 0.47
 @ 3-year 58% (95% CI 43-72) 66% (95% CI 51-80) 0.47
a

All patients received conditioning with the Hopkins regimen (4) which included fludarabine 150 mg/m, cyclophosphamide 29 mg/kg and 200cGy TBI. GvHD prophylaxis was cyclophosphamide 50 mg/kg given on post-transplant days +3 and +4 followed on day +5 with tacrolimus (target level 5-10 ng/mL) or cyclosporine (target level 150-300 ng/mL) and 15 mg/kg mycophenolate every 8 hr not to exceed a total daily dose of 3000 mg. Mycophenolate was discontinued after day +35 and the calcineurin inhibitor (tacrolimus or cyclosporine) on day +180 unless continued for treatment of GvHD.

b

Scored by NIH criteria (11). Previously reported data on chronic GvHD for the haplo-BM cohort (5,6) was scored by the historical Seattle criteria.

In this report, we did not find a significant difference in the rate or global severity of chronic GvHD after haplo-BM or haplo-PB transplantation. Other studies using post-transplant cyclophosphamide as GvHD prophylaxis after ablative or reduced-intensity conditioning and transplantation with either HLA-matched related or unrelated donors have also observed no increase in the incidence or global severity of chronic GvHD after transplantation with PB grafts (12, 13). Regulatory T-cells are thought to play a role in modulating GvHD (14). The fact that regulatory T-cells are resistant to cyclophosphamide (15) may be a possible explanation for the low incidence of severe chronic GvHD with a regimen of GvHD prophylaxis which includes post-transplant cyclophosphamide.

This study is limited by its modest sample size and disease heterogeneity. However, we attempted to adjust for the heterogeneity by matching BM and PB recipients for DRI and patient age. Acknowledging these limitations, we observed comparable overall survival after transplantation of haplo-BM and haplo-PB in the multi-center setting (7, 9). The lower relapse rates observed after transplantation of haplo-PB deserves exploration in a larger cohort of patients with relatively homogenous malignant hematologic disease. Plausible explanations for our observation in the current analysis include differences in diseases between treatment groups and unknown or unmeasured factors that may have influenced relapse. With increasing numbers of haploidentical donor transplantation for hematologic malignancy the question of optimal graft type merit further exploration in larger homogenous datasets.

Acknowledgments

This research was supported, in part, by grants U01-HL069294 from the National Heart, Lung and Blood Institute and the National Cancer Institute, U24-CA76518 from the National Heart, Lung and Blood Institute, the National Cancer Institute and the National Institute of Allergy and Infectious Diseases, CA 18029 from the National Cancer Institute and LLR Funding.

Footnotes

Conflict of interest statement: There are no conflicts of interest to report.

Financial disclosure: The authors have nothing to disclose.

References

  • 1.Stem Cell Trialists' Collaborative Group. Allogeneic peripheral blood stem cell compared with bone marrow transplantation in the management of hematologic malignancies: An individual patient data meta-analysis of nine randomized trials. J Clin Oncol. 2005;23:5074–5087. doi: 10.1200/JCO.2005.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Anasetti C, Logan BR, Lee SJ, Waller EK, Weisdorf DJ, Wingard JR, et al. Peripheral-blood stemm cells versus bone marrow from unrelated donors. N Engl J Med. 2012;367:1487–1496. doi: 10.1056/NEJMoa1203517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Eapen M, Logan BR, Horowitz M, Zhong X, Perales MA, Lee SJ, et al. Bone marrow or peripheral blood for reduced-intensity conditioning unrelated donor transplantation. J Clin Oncol. 2015;33:364–369. doi: 10.1200/JCO.2014.57.2446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Luznik L, O'Donnell PV, Symons HJ, Chen AR, Leffell MS, Zahurak M, et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant. 2008;14:641–50. doi: 10.1016/j.bbmt.2008.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Brunstein CG, Fuchs EJ, Carter SL, Karanes C, Costa L, Wu J, et al. Alternative donor transplantation after reduced intensity conditioning: results of parallel phase 2 trials using partially HLA-mismatched related bone marrow or unrelated double umbilical cord blood grafts. Blood. 2011;118:282–8. doi: 10.1182/blood-2011-03-344853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Eapen M, O'Donnell P, Brunstein C, Wu J, Barowski K, Mendizabal A, et al. Mismatched related and unrelated donors for allogeneic hematopoietic cell transplantation for adults with hematologic malignancies. Biol Blood Marrow Transplant. 2014;20:1485–1492. doi: 10.1016/j.bbmt.2014.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Castagna L, Crocchiolo R, Furst S, Bramanti S, El Cheikh J, Sarina B, et al. Bone marrow compared with peripheral blood stem cells for haploidentical transplantation with a nonmyeloablative conditioning regimen and post-transplatation cyclophosphamide. Biol Blood Marrow Transplant. 2014;20:724–729. doi: 10.1016/j.bbmt.2014.02.001. [DOI] [PubMed] [Google Scholar]
  • 8.Raj K, Pagliuca A, Bradstock K, Noriega V, Potter V, Streetly M, et al. Peripheral blood hematopoietic stem cells for transplantation of hematological diseases from related, haploidentical donors after reduced-intensity conditioning. Biol Blood Marrow Transplant. 2014;20:881–903. doi: 10.1016/j.bbmt.2014.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bradstock K, Bilmon I, Kwan J, Blyth E, Micklethwaite K, Huang G, et al. Influence of stem cell source on outcomes of allogeneic reduced-intensity conditioning therapy transplants using haploidentical related donors. Biol Blood Marrow Transplant. 2015;21:1641–1645. doi: 10.1016/j.bbmt.2015.06.006. [DOI] [PubMed] [Google Scholar]
  • 10.Armand P, Haesook TK, Logan BR, Wang Z, Alyea EP, Kalaycio ME, et al. Validation and refinement of the Disease Risk Index for allogeneic stem cell transplantation. Blood. 2014;123:3664–3671. doi: 10.1182/blood-2014-01-552984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Filipovich AH, Weisdorf D, Pavletic S, Socie G, Wingard JR, Lee SJ, 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 and Marrow Transplant. 2005;11:945–956. doi: 10.1016/j.bbmt.2005.09.004. [DOI] [PubMed] [Google Scholar]
  • 12.Mielcarek M, Furlong T, O'Donnell PV, Storer BE, McCune JS, Storb R, et al. Posttransplantation cyclophosphamide for prevention of graft-versus-host disease after HLA-matched mobilized blood cell transplantation. Blood. 2016;127:1502–1508. doi: 10.1182/blood-2015-10-672071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Moiseev IS, Pirogova OV, Alyanski L, Babenko EV, Gindina TL, Darskaya EI, et al. Graft-versus-host disease prophylaxis in unrelated peripheral blood stem cell transplantation with post-transplantation cyclophosphamide, tacrolimus and mycophenolate mofetil. Biol Blood Marrow Transpl. 2016 doi: 10.1016/j.bbmt.2016.03.004. in press. [DOI] [PubMed] [Google Scholar]
  • 14.Ganguly S, Ross DB, Panoskaltsis-Mortari A, Kanakry C, Blazar BR, Levy RB, et al. Donor CD4+ Foxp3+ regulatory T cells are necessary for posttransplantation cyclophosphamide-mediated protection against GVHD in mice. Blood. 2014;124:2131–2141. doi: 10.1182/blood-2013-10-525873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kanakry CG, Ganguly S, Zahurak M, Bolanos-Meade J, Thoburn C, Perkins B, et al. Aldehyde dehydrogenase expression drives human regulatory T cell resistance to posttransplantation cyclophosphamide. Sci Transl Med. 2013;5:1–12. doi: 10.1126/scitranslmed.3006960. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES