Toxicities of high-dose conditioning regimens have limited the use of conventional unrelated donor hematopoietic cell transplantation (HCT) to younger, medically fit patients. Based on preclinical studies, an HCT approach has been developed for elderly or medically infirm patients with HLA-matched or mismatched unrelated donors. In this study, 52 patients with hematological diseases were included. Most (88%) had preceding unsuccessful conventional HCT or refractory/advanced disease. Patients were treated with fludarabine 30 mg/m2/d from days −4 to −2, 2 Gy total body irradiation on day 0, cyclosporine at 6.25 mg/kg twice daily from day −3, and mycophenolate mofetil at 15 mg/kg twice daily from day 0. Durable donor chimerism was attained in 88% of the patients. By day 28, a median of 100% of CD56+ cells were of donor origin. Granulocyte and T-cell donor chimerism increased to medians of 100% on day 56 and day 180 (range, 55%-100%), respectively. Acute GVHD, grade II, was seen in 42% (CI, 29%-56%); grade III in 8% (CI, 0%-15%); and grade IV in 13% (CI, 4%-23%) of patients; it was fatal in 9%. The 100-day transplantation-related mortality was 11%. Complete remissions, including molecular remissions, were seen in 45% of patients with measurable disease before transplantation. Mortality from disease progression was 27% at one year. With a median follow-up of 19 months, 18 of the 52 patients (35%) were alive and 25% were in remission. HCT from HLA-matched or mismatched unrelated donors can be performed with a reduced intensity conditioning regimen in patients ineligible for conventional HCT.

Conventional allogeneic unrelated hematopoietic cell transplantation (HCT) for patients with hematological malignancies involves conditioning with high doses of systemic chemo/radiation therapy.1-3 Regimen-related toxicities have limited the procedure to medically fit patients generally no older than 50 years, with therapy administered on specialized hospital wards. Given that median ages of patients with chronic myelocytic leukemia (CML), acute myelocytic leukemia (AML), chronic lymphocytic leukemia (CLL), multiple myeloma (MM), myelodysplastic syndromes (MDS), and non-Hodgkin lymphoma (NHL) range from 65-70 years, most patients with these diseases are not candidates for conventional unrelated HCT.4 In an attempt to reduce morbidity and mortality associated with allogeneic HCT in elderly patients, reduced-intensity regimens have been developed with the aim to obtain donor engraftment and use graft-versus-tumor effects to eradicate underlying malignancies.5-12 In these protocols, donor engraftment is achieved with regimens conveying different degrees of myelosuppression ranging from minimal to severe. Based on preclinical studies in the canine model, we developed an allogeneic HCT approach with minimal hematopoietic and overall toxicities, in which the burden of tumor eradication has been shifted from cytotoxic therapy to graft-versus-tumor effects.11,13The conditioning regimen consists of 2 Gy total body irradiation (TBI), and both engraftment and graft-versus-host disease (GVHD) are controlled with postgrafting immunosuppression using mycophenolate mofetil (MMF) combined with cyclosporine (CSP). Early results with HLA-identical sibling grafts in elderly or medically infirm patients with hematological malignancies using this regimen have been encouraging, and remissions, including molecular remissions, have been accomplished in more than half of patients receiving transplants.11 Here, we present data extending this approach to patients with HLA-matched and mismatched unrelated donors. In contrast to the transplants with related grafts, patients were given 3 doses of fludarabine in addition to 2 Gy TBI to assure engraftment.

Patients

Between October 26, 1998, and July 14, 2000, 52 consecutive patients were treated at the University of Leipzig, Fred Hutchinson Cancer Research Center/University of Washington Medical Center and Veteran's Administrations Medical Center in Seattle, WA, Stanford University, CA, and the University of Colorado, CO. Results were analyzed as of September 1, 2001.

Included in this study were patients with hematological diseases treatable by allogeneic HCT who were ineligible for conventional allogeneic HCT because of age (older than 50 years) and/or concomitant diseases (eg, liver cirrhosis, Shwachman-Diamond syndrome) or preceding extensive therapies, such as failed autologous or allogeneic HCT. Exclusion criteria were creatinine clearance ≤ 50 mL/min, cardiac ejection fraction ≤ 30%, bilirubin > 2 × and/or transaminases > 4 × upper limit of normal, diffusing lung carbon monoxide (DLCO) < 35%, and Karnofsky performance score < 50.

Patient characteristics are summarized in Table1. Underlying diseases were MDS (n = 8), including 2 patients evolved to AML, AML (n = 10), acute lymphoblastic leukemia (ALL) (n = 2), CML (n = 12), CLL (n = 3), NHL (n = 6), HD (n = 1), MM (n = 8), Waldenström macroglobulinemia (WM, n = 1), and paroxysmal nocturnal hemoglobinuria (PNH, n = 1). Most patients (n = 46; 88%) were high-risk candidates as defined by relapse after preceding HCT or by refractory/advanced disease. All other patients were considered low risk (n = 6). Overall, 22 patients (42%) had failed at least one previous autologous or allogeneic HCT. Median patient age was 48 years (range, 6-65 years), and median donor age was 35 years (range, 19-58 years). The study protocol was approved by the Institutional Review Boards of the participating institutions (University Hospital, Leipzig, Germany; Fred Hutchinson Cancer Research Center, Seattle, WA; University Hospital Stanford, CA; and University of Colorado, Denver) and written informed consents approved by the Institutional Review Boards were obtained from all patients.

Table 1.

Characteristics of patients and grafts (grouped by diagnoses)

UPNAge (y)/sexDisease + statusTherapy before HCTContraindications to conventional HCTMos. from Dx to HCTSource of HSCCD34+ cells × 106/kgCD3+ cells × 108/kgHLA mismatch
Rejection vectorGVHD vector
SPN2292 21/M ALL/CR2 3x Dnr/Vcr/Asp/Pred, MTX, Flu/Ara-C/Ida Hepatic dysfunction, aspergillosis sinuses 64 Blood 7.1 1.2 1-153 1-153 
UL755 27/M ALL/CR2 1x Cy/Dnr/Asp/Vcr, 2x HAM, purinetol/MTX S.p. ventilation for int. pneumonia + septicemia 59 Blood 6.1 5.3 C C 
SPN2252 57/F AML/CR1 2x Ida/Ara-C Age Marrow 3.5 0.4 1-153 1-153 
UL676 45/M AML/CR2 1x Ida/Ara-C, 1x Mito-FLAG Autologous HCT 15 Blood 5.8 3.1 DQBI DQBI 
UL652 59/M AML/CR3 4x Ida/Ara-C, 2x Mito-FLAG, 1x TBI/Ara-C, VP16 Age + autologous HCT 27 Blood 9.6 2.1 A* A* 
FH15303 59/F AML/CR3 3x Ida/Ara-C, 2x Mito/VP16, Topo/Ara-C, i.t. MTX, craniospinal RT Age 21 Blood 2.6 3.8 1-153 1-153 
UL637 53/M AML/CR3 3x Ida/Ara-C, Ara-C/Mito/Ida/VP16, 1x Mito/FLAG Age + aspergillosis + liver cirrhosis 36 Blood 4.7 5.2 C*  
FH16411 40/F AML/CR4 4x Ida/Ara-C, 2x Ara-C, HD-Cy, i.t. MTX, craniospinal RT Suspected aspergillosis + hepatic dysfunction 21 Blood 16.8 5.5 1-153 1-153 
UL665 51/F AML/PR2 3x Ida/Ara-C, 2x Mito-FLAG Age + s.p. ventilation for int. pneumonia + septicemia 35 Blood 5.00 3.7 1-153 1-153 
UL668 61/M AML/progression 4x Ida/Ara-C, 2x Mito/VP16, 3x Mito/FLAG, HD-BuCy/VP16, CHOP, vindesine Age + autologous HCT 82 Blood 2.87 2.09 1-153 1-153 
UL680 60/F AML/progression 1x ICE, 1x HAM, 1x Mito-FLAG, 1x HAM Age 21 Blood 3.3 1.39 A*BC* A*BC* 
UL692 48/F AML/rel1 1x Ida/Ara-C, 3x Mito/Ara-C Age + Karnofsky 60 12 Blood 14.3 3.7 B B 
UL656 47/M CLL/PR HD-chlorambucil, 6x Flu/Cy Age + patient decision Blood 21.6 4.5 1-153 1-153 
UL698 50/M CLL/PR 6x CHOP, 2x Flu/Cy, 1x Dexa-BEAM Age + s.p. recurrent septicemias 77 Blood 5.5 3.4 1-153 1-153 
FH16078 59/M CLL/rel Chlorambucil, 14x Flu, 6x Flu/Cy Age 78 Blood 12.80 2.27 1-153 1-153 
UL597 48/M CML/AP Hu/IFN, 1x Ida/Ara-C, HD-Bu, Hu/IFN Autologous HCT 41 Blood 10.3 4.6 C* C* 
UL641 45/F CML/AP 1x Ida/Ara-C, IFN/Hu, IFN/Ara-C Adipositas, hypertension 30 Blood 3.2 2.6   
UL670 47/M CML/AP Hu, Ida/Ara-C, Hu/IFN, HD-Bu, Hu/IFN Autologous HCT 42 Marrow 1.7 0.22 
UL672 50/F CML/CP 1x Ida/Ara-C, IFN/Hu Age 32 Blood 3.9 1-153 1-153 
UL695 46/M CML/AP Hu, IFN, Ida/Ara-C, HD-Bu Autologous HCT 63 Blood 14.2 3.8 A A 
UL709 62/M CML/CP Hu/IFN, Ara-C Age 24 Blood 5.3 6.2 1-153 1-153 
UL725 57/M CML/CP Hu, IFN, Hu Age 80 Blood 4.5 1-153 1-153 
UL730 57/M CML/CP Hu/IFN Age 75 Blood 5.8 3.3 C* A C* 
         DQB1* DQB1* 
UL735 57/F CML/CP Hu, Ida/Ara-C, IFN/Hu, HD-Bu, Ara-C/IFN, Hu, Tg Age + autologous HCT 26 Blood 5.5 10 1-153 1-153 
UL754 54/M CML/CP2 Hu, IFN, Ida/Ara-C, HD-Bu, IFN, Hu, Vcr/Pred, ST1571, Ida/Ara-C Age + autologous HCT 74 Blood 6.1 3.4 C*
DRB1
DQB1* 
C*
DRB1 
UC001 36/F CML/AP Hu/IFN, BU/mel, Ida/Ara-C/DLI, Flu/ATG/TLI, FLAG Previous HLA-id sibling HCT 24 Marrow 2.76 0.45   
UC002 33/M CML/BC IFN/Hu, Cy/TBI, Mito/Flu/Ara-C, Mylotarg Previous HLA-matched unrelated HCT 52 Marrow 1.88 0.29   
FH15764 43/M Hodgkin disease/rel 3x ABVD, 3x Chl-VPP, Dnr, Vbl, 6x VP16, RT pelvic nodes, L hemipelvis/groin, BEAC Autologous HCT 32 Blood 5.65 1.54 1-153 1-153 
UL701 40/M NHL/refr B-cell 8x CHOP, Vbl/Dexa, Dexa-BEAM, HD-MTX, HD-BEAM, Bu/Thiotepa 2x Autologous HCT 21 Blood 6.2 4.9 A* A* 
SPN1463 45/M NHL/rel diff. large cell MOPP/ABV, radiotherapy, 2x DHAP, HD-Cy, Vcr, VP16, Mito/Mel, Rituxan, Cy/Flu, Taxol Autologous HCT 41 Blood 3.60 1.7 C* 1-153 
FH13715 42/M NHL/rel MCL 6x CHOP, Cy/VP16, 2x Gemcitabine, 2x MINE/Rituxan Autologous HCT 63 Marrow ND ND 1-153 1-153 
FH16020 46/M NHL/rel MCL 6x CHOP, Rituxan, Cy/VP16/BCNU Autologous HCT 45 Blood 8.22 3.83 1-153 1-153 
UL744 62/M NHL/rel high grade T cell 4x CHOP, 2x Dexa-BEAM, HD-BEAM, Cy/Pred, HAM, Dexa-BEAM Age + autologous HCT 29 Marrow 3.3 0.6 BC* BC* 
FH15780 62/F NHL/PR MCL 4x CNOP, 2x Mito/Flu, Flu, Rituxan, ESHAP, Cy/VP16, 3x Topo/Taxol Age 41 Marrow 1.17 ND  1-153 1-153 
FH17028 52/M WM/refr Chlorambucil, 14x Flu, Cladribine, Rituxan Age 184 Marrow 2.19 ND  1-153 1-153 
FH15390 6/M MDS/RA No prev tx Shwachman-Diamond syndrome Marrow ND ND 1-153 1-153 
SPN2219 41/M MDS/CMML Pred, Hu Congestive heart failure and performance status Blood 12.30 2.00 1-153 1-153 
SPN2269 39/M MDS/RAEB-T No prev tx Poor performance status, recurrent infections, type II diabetes Blood 7.4 0.8 1-153 1-153 
UL760 33/F MDS/CMML 2x Ida/Ara-C Aspergillosis Blood 7.9 5.1 BC* BC* 
FH11799 55/F MDS/RAEB No prev tx Age 165 Marrow 4.07 0.3 1-153 1-153 
FH15608 40/F MDS/AML/CR 3x Ida/Ara-C Reflex sympathetic dystrophy, chr. foot ulcer, chr. pain syndrome 15 Blood 1.48 1.47 1-153 1-153 
FH16391 46/M MDS/AML refr 6x CVAP/Bleomycin, Cy/VP16, BEAC, Ara-C/Topo Autologous HCT Marrow 2.49 0.17 1-153 1-153 
FH15595 65/F MDS/MPS Hu Age 11 Blood 2.21 3.08 1-153 1-153 
UL664 40/M MM/rel 10x Vcr/Cy/Mel, Cy, 3x VAD, TBI/Mel, HD-Mel, 2x Benda/Pred 2x Autologous HCT 109 Marrow 2.7 0.6 C*
DQB1* 
C* DQB1* 
UL714 59/M MM/rel 9x Mel, 2x VAD, 2x HD-Cy, HD-Dexa, 1x Mel Age 56 Blood 4.1 4.3 1-153 C* 
UL733 48/F MM/rel Dexa, 2x VP16/Ifo, TBI/Mel, 2x Benda Autologous HCT 73 Blood 21.1 8.5 1-153 1-153 
UL747 49/M MM/PR 13x Mel/Pred, 3x VAD, 1x HD-Cy, 2x HD-Mel, 3x Thalid/Cy/VP16/Dexa, Thalid 2x Autologous HCT 65 Marrow 5.2 0.4 1-153 1-153 
UL753 50/M MM/PR 6x VAD Age 21 Blood 15.6 3.9 C* C* 
FH15711 41/M MM/refr VAD, DCEP/Thalid, CAD, Dexa/Thalid, Dexa Patient's request 31 Blood 5.50 2.27 1-153 1-153 
UL765 61/M MM/PR 1x Mel/Pred, 3x Ifo/Epi, 1x HD-Mel, IFN, 1x Ifo/Epi, 6x Benda/Vbl, IFN Age + autologous HCT 66 Blood 6.3 4.3 AC* AC* 
FH14886 61/M MM/refr 3x VAD, HD-Mel Age + autologous HCT 30 Blood 7.43 1-153 1-153 
UL703 30/M PNH CSP, Pred Sarcoidosis 15 Blood 5.6 3.2 C* 1-153 
UPNAge (y)/sexDisease + statusTherapy before HCTContraindications to conventional HCTMos. from Dx to HCTSource of HSCCD34+ cells × 106/kgCD3+ cells × 108/kgHLA mismatch
Rejection vectorGVHD vector
SPN2292 21/M ALL/CR2 3x Dnr/Vcr/Asp/Pred, MTX, Flu/Ara-C/Ida Hepatic dysfunction, aspergillosis sinuses 64 Blood 7.1 1.2 1-153 1-153 
UL755 27/M ALL/CR2 1x Cy/Dnr/Asp/Vcr, 2x HAM, purinetol/MTX S.p. ventilation for int. pneumonia + septicemia 59 Blood 6.1 5.3 C C 
SPN2252 57/F AML/CR1 2x Ida/Ara-C Age Marrow 3.5 0.4 1-153 1-153 
UL676 45/M AML/CR2 1x Ida/Ara-C, 1x Mito-FLAG Autologous HCT 15 Blood 5.8 3.1 DQBI DQBI 
UL652 59/M AML/CR3 4x Ida/Ara-C, 2x Mito-FLAG, 1x TBI/Ara-C, VP16 Age + autologous HCT 27 Blood 9.6 2.1 A* A* 
FH15303 59/F AML/CR3 3x Ida/Ara-C, 2x Mito/VP16, Topo/Ara-C, i.t. MTX, craniospinal RT Age 21 Blood 2.6 3.8 1-153 1-153 
UL637 53/M AML/CR3 3x Ida/Ara-C, Ara-C/Mito/Ida/VP16, 1x Mito/FLAG Age + aspergillosis + liver cirrhosis 36 Blood 4.7 5.2 C*  
FH16411 40/F AML/CR4 4x Ida/Ara-C, 2x Ara-C, HD-Cy, i.t. MTX, craniospinal RT Suspected aspergillosis + hepatic dysfunction 21 Blood 16.8 5.5 1-153 1-153 
UL665 51/F AML/PR2 3x Ida/Ara-C, 2x Mito-FLAG Age + s.p. ventilation for int. pneumonia + septicemia 35 Blood 5.00 3.7 1-153 1-153 
UL668 61/M AML/progression 4x Ida/Ara-C, 2x Mito/VP16, 3x Mito/FLAG, HD-BuCy/VP16, CHOP, vindesine Age + autologous HCT 82 Blood 2.87 2.09 1-153 1-153 
UL680 60/F AML/progression 1x ICE, 1x HAM, 1x Mito-FLAG, 1x HAM Age 21 Blood 3.3 1.39 A*BC* A*BC* 
UL692 48/F AML/rel1 1x Ida/Ara-C, 3x Mito/Ara-C Age + Karnofsky 60 12 Blood 14.3 3.7 B B 
UL656 47/M CLL/PR HD-chlorambucil, 6x Flu/Cy Age + patient decision Blood 21.6 4.5 1-153 1-153 
UL698 50/M CLL/PR 6x CHOP, 2x Flu/Cy, 1x Dexa-BEAM Age + s.p. recurrent septicemias 77 Blood 5.5 3.4 1-153 1-153 
FH16078 59/M CLL/rel Chlorambucil, 14x Flu, 6x Flu/Cy Age 78 Blood 12.80 2.27 1-153 1-153 
UL597 48/M CML/AP Hu/IFN, 1x Ida/Ara-C, HD-Bu, Hu/IFN Autologous HCT 41 Blood 10.3 4.6 C* C* 
UL641 45/F CML/AP 1x Ida/Ara-C, IFN/Hu, IFN/Ara-C Adipositas, hypertension 30 Blood 3.2 2.6   
UL670 47/M CML/AP Hu, Ida/Ara-C, Hu/IFN, HD-Bu, Hu/IFN Autologous HCT 42 Marrow 1.7 0.22 
UL672 50/F CML/CP 1x Ida/Ara-C, IFN/Hu Age 32 Blood 3.9 1-153 1-153 
UL695 46/M CML/AP Hu, IFN, Ida/Ara-C, HD-Bu Autologous HCT 63 Blood 14.2 3.8 A A 
UL709 62/M CML/CP Hu/IFN, Ara-C Age 24 Blood 5.3 6.2 1-153 1-153 
UL725 57/M CML/CP Hu, IFN, Hu Age 80 Blood 4.5 1-153 1-153 
UL730 57/M CML/CP Hu/IFN Age 75 Blood 5.8 3.3 C* A C* 
         DQB1* DQB1* 
UL735 57/F CML/CP Hu, Ida/Ara-C, IFN/Hu, HD-Bu, Ara-C/IFN, Hu, Tg Age + autologous HCT 26 Blood 5.5 10 1-153 1-153 
UL754 54/M CML/CP2 Hu, IFN, Ida/Ara-C, HD-Bu, IFN, Hu, Vcr/Pred, ST1571, Ida/Ara-C Age + autologous HCT 74 Blood 6.1 3.4 C*
DRB1
DQB1* 
C*
DRB1 
UC001 36/F CML/AP Hu/IFN, BU/mel, Ida/Ara-C/DLI, Flu/ATG/TLI, FLAG Previous HLA-id sibling HCT 24 Marrow 2.76 0.45   
UC002 33/M CML/BC IFN/Hu, Cy/TBI, Mito/Flu/Ara-C, Mylotarg Previous HLA-matched unrelated HCT 52 Marrow 1.88 0.29   
FH15764 43/M Hodgkin disease/rel 3x ABVD, 3x Chl-VPP, Dnr, Vbl, 6x VP16, RT pelvic nodes, L hemipelvis/groin, BEAC Autologous HCT 32 Blood 5.65 1.54 1-153 1-153 
UL701 40/M NHL/refr B-cell 8x CHOP, Vbl/Dexa, Dexa-BEAM, HD-MTX, HD-BEAM, Bu/Thiotepa 2x Autologous HCT 21 Blood 6.2 4.9 A* A* 
SPN1463 45/M NHL/rel diff. large cell MOPP/ABV, radiotherapy, 2x DHAP, HD-Cy, Vcr, VP16, Mito/Mel, Rituxan, Cy/Flu, Taxol Autologous HCT 41 Blood 3.60 1.7 C* 1-153 
FH13715 42/M NHL/rel MCL 6x CHOP, Cy/VP16, 2x Gemcitabine, 2x MINE/Rituxan Autologous HCT 63 Marrow ND ND 1-153 1-153 
FH16020 46/M NHL/rel MCL 6x CHOP, Rituxan, Cy/VP16/BCNU Autologous HCT 45 Blood 8.22 3.83 1-153 1-153 
UL744 62/M NHL/rel high grade T cell 4x CHOP, 2x Dexa-BEAM, HD-BEAM, Cy/Pred, HAM, Dexa-BEAM Age + autologous HCT 29 Marrow 3.3 0.6 BC* BC* 
FH15780 62/F NHL/PR MCL 4x CNOP, 2x Mito/Flu, Flu, Rituxan, ESHAP, Cy/VP16, 3x Topo/Taxol Age 41 Marrow 1.17 ND  1-153 1-153 
FH17028 52/M WM/refr Chlorambucil, 14x Flu, Cladribine, Rituxan Age 184 Marrow 2.19 ND  1-153 1-153 
FH15390 6/M MDS/RA No prev tx Shwachman-Diamond syndrome Marrow ND ND 1-153 1-153 
SPN2219 41/M MDS/CMML Pred, Hu Congestive heart failure and performance status Blood 12.30 2.00 1-153 1-153 
SPN2269 39/M MDS/RAEB-T No prev tx Poor performance status, recurrent infections, type II diabetes Blood 7.4 0.8 1-153 1-153 
UL760 33/F MDS/CMML 2x Ida/Ara-C Aspergillosis Blood 7.9 5.1 BC* BC* 
FH11799 55/F MDS/RAEB No prev tx Age 165 Marrow 4.07 0.3 1-153 1-153 
FH15608 40/F MDS/AML/CR 3x Ida/Ara-C Reflex sympathetic dystrophy, chr. foot ulcer, chr. pain syndrome 15 Blood 1.48 1.47 1-153 1-153 
FH16391 46/M MDS/AML refr 6x CVAP/Bleomycin, Cy/VP16, BEAC, Ara-C/Topo Autologous HCT Marrow 2.49 0.17 1-153 1-153 
FH15595 65/F MDS/MPS Hu Age 11 Blood 2.21 3.08 1-153 1-153 
UL664 40/M MM/rel 10x Vcr/Cy/Mel, Cy, 3x VAD, TBI/Mel, HD-Mel, 2x Benda/Pred 2x Autologous HCT 109 Marrow 2.7 0.6 C*
DQB1* 
C* DQB1* 
UL714 59/M MM/rel 9x Mel, 2x VAD, 2x HD-Cy, HD-Dexa, 1x Mel Age 56 Blood 4.1 4.3 1-153 C* 
UL733 48/F MM/rel Dexa, 2x VP16/Ifo, TBI/Mel, 2x Benda Autologous HCT 73 Blood 21.1 8.5 1-153 1-153 
UL747 49/M MM/PR 13x Mel/Pred, 3x VAD, 1x HD-Cy, 2x HD-Mel, 3x Thalid/Cy/VP16/Dexa, Thalid 2x Autologous HCT 65 Marrow 5.2 0.4 1-153 1-153 
UL753 50/M MM/PR 6x VAD Age 21 Blood 15.6 3.9 C* C* 
FH15711 41/M MM/refr VAD, DCEP/Thalid, CAD, Dexa/Thalid, Dexa Patient's request 31 Blood 5.50 2.27 1-153 1-153 
UL765 61/M MM/PR 1x Mel/Pred, 3x Ifo/Epi, 1x HD-Mel, IFN, 1x Ifo/Epi, 6x Benda/Vbl, IFN Age + autologous HCT 66 Blood 6.3 4.3 AC* AC* 
FH14886 61/M MM/refr 3x VAD, HD-Mel Age + autologous HCT 30 Blood 7.43 1-153 1-153 
UL703 30/M PNH CSP, Pred Sarcoidosis 15 Blood 5.6 3.2 C* 1-153 
*

Antigen-level mismatch.

Allele-level mismatch.

Antigen match.

F1-153

Allele match.

Forty-three patients were mismatched in addition at DPB1 for either one or both alleles by rejection vector (n = 43) or by GVHD vector (n = 37).

ND indicates not done.

HLA typing

The 52 donor-recipient pairs were selected on the basis of serological matching for HLA-A, B, and C, molecular matching for HLA-DRB1, and in 22 patients for HLA-DQB1. Of donor-recipient pairs, 15 (29%) were HLA mismatched, and 37 (71%) were matched at the antigen level. Retrospective allele level typing was performed on 49 of the 52 donor-recipient pairs for HLA-A, B, C, DQB1, and DPB1 alleles using direct automated fluorescent methods as previously described.14,15 Of the 15 pairs with antigen mismatches, 6 were found to encode additional allele mismatches, and 5 of the 37 HLA antigen-matched pairs were found to be mismatched for one HLA allele. Overall, one or more antigen and/or allele level mismatches were recognized in 20 of the 52 pairs (38%; Table 1). Mismatches by rejection vector (host-versus-graft direction) were present in 19 pairs, including one (n = 12), 2 (n = 6), and 3 (n = 1) mismatches and by GVHD vector (graft-versus-host direction) in 17 pairs including one (n = 9), 2 (n = 6), and 3 (n = 2) mismatches. Results of these studies are shown in Table 1.

Treatment and evaluations

Patients were treated with 3 doses of fludarabine, 30 mg/m2/d, from days −4 to −2 and a single fraction of 2 Gy total body irradiation (TBI) delivered at 0.07 Gy/min from linear accelerators on day 0. In the afternoon of day 0, donor hematopoietic cells were infused. In 39 patients, the source of donor hematopoietic cells were granulocyte colony–stimulating factor (G-CSF)–stimulated peripheral blood cells (PBCs) containing medians of 6.0 × 106 (range, 1.5 × 106-21.6 × 106) CD34+and 3.4 × 108 (range, 0.8 × 108-10.0 × 108) CD3+cells/kg. In 13 patients marrow grafts were infused containing medians of 2.7 × 106 (range, 1.2 × 106-5.2 × 106) CD34+and 0.3 × 108 (range, 0.2 × 108-0.6 × 108) CD3+cells/kg. CSP was administered orally at 6.25 mg/kg twice daily from day −3. CSP levels were targeted to the upper therapeutic range (about 500 ng/mL; Abbott TDX, Abbott Park, IL) until day +35 and then discontinued (one patient). In the subsequent 10 patients, CSP was tapered from day 64 after transplantation until day 180. In the remaining 41 patients, CSP was tapered starting on day 100 through day 180. MMF was given orally at 15 mg/kg beginning in the afternoon of day 0 and then twice daily to day +27 (first 13 patients). In the remaining 39 patients, MMF was given in full doses to day +40 and then tapered through day +96.

Standard prophylaxis against Pneumocystis carinii, fungal infections, toxoplasmosis, and cytomegalovirus (CMV) infections was used.16-18 Patients with chronic GVHD requiring systemic immunosuppressive therapy continued prophylaxis against P carinii and pneumococcal infections. For outpatient transplantations, scheduled follow-up included 2 to 3 clinic visits per week for the first month, and then once or twice weekly or as clinically indicated thereafter.

The degrees of donor chimerism among peripheral blood T cells, granulocytes, and nucleated marrow cells were assessed at days 28, 56, 84, 180, and 360 after HCT using fluorescence in situ hybridization to detect X and Y chromosomes for recipients of grafts from sex-mismatched donors, and polymerase chain reaction (PCR)–based analyses of polymorphic microsatellite regions for recipients of sex-matched transplants.19 In patients who received transplants at the University of Leipzig, donor chimerism was also evaluated on day 14 after HCT.

The primary study end point was mixed chimerism on day 28, defined as between 5% and 95% peripheral blood donor T cells. Disease evaluations were performed monthly. Donor lymphocyte infusions (DLIs) were given to 6 patients for relapse/disease progression between 57 and 382 (median, 135) days after HCT (median, 10 × 106/kg CD3+ cells infused). Of the 6 patients, 4 received a second dose DLI (median, 43 × 106/kg CD3+cells).

Acute and chronic GVHD were graded as described.20,21Disease responses were assessed using standard criteria. PCR-based techniques were used to test for bcr-abl transcripts (CML),22 clonal immunoglobulin rearrangements (lymphoid malignancies), or other chromosomal translocations. Toxicities were determined using the Common Toxicity Criteria, Version 2.0.

Statistical analyses

Survival and progression-free survival were estimated by the Kaplan-Meier method. Cumulative incidence estimates were calculated for acute GVHD, relapse, and mortality from various causes.23Risk factors for acute GVHD, rejection, relapse, and survival were analyzed using proportional hazards regression models, treating death prior to acute GVHD, rejection, or relapse as a competing event. Rejection was treated as a competing event for analysis of acute GVHD and as a time-dependent covariate for analyses of death and relapse. The factors considered for survival, TRM, and relapse were disease risk, diagnosis, Class I and Class II mismatch, and rejection. The multivariate models were constructed in a stepwise fashion, and no more than 2 variables ever entered any of the models. All Pvalues were derived from likelihood ratio statistics and were 2-sided.

Peripheral blood cell changes, allogeneic engraftment, and graft rejection

Figure 1 summarizes peripheral blood neutrophil and platelet changes to day +30 after HCT. A median of 4 units (range, 0-166 units) of platelets and a median of 8 units (range, 0-34 units) of red blood cells were transfused. Nine patients (17%) did not require any transfusions, 16 did not require platelet transfusions, and 9 did not require red blood cell transfusions. No adverse hemolytic reactions occurred in 32 blood group (18 minor and 14 major) incompatible transplantations. Median hospital stay for 22 patients treated in the US and eligible for outpatient therapy was 16 days (range, 0 to 56 days), while the 30 patients who received transplants in Germany and were ineligible for outpatient treatment had a median hospital stay of 40 days (range, 13 to 122 days).

Fig. 1.

Engraftment after unrelated HCT.

Neutrophil and platelet changes after HCT. Graphs show the medians (solid lines) and ranges (broken lines) of neutrophil and platelet counts of the 52 patients from day 0 through day 30. ○ indicates the minimum and maximum values on each day.

Fig. 1.

Engraftment after unrelated HCT.

Neutrophil and platelet changes after HCT. Graphs show the medians (solid lines) and ranges (broken lines) of neutrophil and platelet counts of the 52 patients from day 0 through day 30. ○ indicates the minimum and maximum values on each day.

Close modal

Complete donor chimerism (100% donor cells) was generally attained by day 28 for natural killer (NK) cells, by day 56 for granulocytes, and by day 180 for CD3+ cells. Figure2 illustrates the median percentages of peripheral blood donor CD3+ cells, CD56+ cells, and granulocytes through day 180, and Table2 shows the percentages of donor CD3 cells on days 28 and 56 for each individual patient.

Fig. 2.

Percent donor chimerism after unrelated HCT.

Median percentages of donor peripheral blood CD56+ cells (NK-cells, ▵), CD3+ cells (T-lymphocytes; ○), and CD15+ cells (granulocytes; ●) are shown for the first 180 days after transplantation. The numbers of patients analyzed at days 28, 56, 84, and 180 were, respectively, 45, 40, 37, and 27 for T cells; 46, 40, 38, and 26 for granulocytes; and 27, 21, 19, and 15 for NK cells.

Fig. 2.

Percent donor chimerism after unrelated HCT.

Median percentages of donor peripheral blood CD56+ cells (NK-cells, ▵), CD3+ cells (T-lymphocytes; ○), and CD15+ cells (granulocytes; ●) are shown for the first 180 days after transplantation. The numbers of patients analyzed at days 28, 56, 84, and 180 were, respectively, 45, 40, 37, and 27 for T cells; 46, 40, 38, and 26 for granulocytes; and 27, 21, 19, and 15 for NK cells.

Close modal
Table 2.

Outcomes after unrelated HCT (grouped by diagnoses)

UPNDisease + status% Donor CD3 cellsGraft rejectionGVHDSurvival, dSummary of outcome as of 09/01/2001
Day 28Day 56Acute (grade)Chronic
SPN2292 ALL/CR2 90 100 No II Ext 211 CR, cGVHD, pneumonia/MOF 
UL755 ALL/CR2 83 88 No IV No 109 CR, aGVHD, infection 
SPN2292 AML/CR1 100 65 No II Lim > 549 CR, alive 
UL676 AML/CR2 85 100 No II Lim 156 CR, sudden death 
UL652 AML/CR3 100 100 No Lim > 844 CR, alive 
FH15303 AML/CR3 90 90 No III No 144 CR, GI bleed 
UL637 AML/CR3 100 100 No IV No 170 CR, aGVHD 
FH16411 AML/CR4 100 100 No II Ext 401 CR, CNS infection/sev. GvHD 
UL665 AML/PR2 75 80 No II Lim > 789 CR, alive 
UL668 AML/prog 96 99 No II Ext 624 CR, infection (urosepsis) 
UL680 AML/prog 95 99 No II Lim 218 Relapse 
UL692 AML/rel1 100 100 No II Lim > 705 CR, alive 
UL656 CLL/PR 100 100 No Lim > 817 CR, alive 
UL698 CLL/PR 88 86 No II Ext 482 CR, suicide, chronic GVHD 
FH16078 CLL/rel 99 99 No II Ext > 480 CR, alive 
UL597 CML/AP 75 80 No III No 122 Progression 
UL641 CML/AP 70 NA No II No 46 NE, TTP 
UL670 CML/AP NA Yes NA NA 172 Relapse 
UL672 CML/CP 75 85 No III Lim 546 CR, cGVHD 
UL695 CML/AP 100 100 No IV Lim > 698 CR, alive 
UL709 CML/CP 80 70 No IV Lim 312 CR, cGVHD 
UL725 CML/CP NA Yes NA NA 203 NE, infection 2nd BMT 
UL730 CML/CP 90 93 No IV No 61 NE, aGVHD 
UL735 CML/CP 95 100 No No 377 Relapse 
UL754 CML/CP2 90 100 No No > 492 CR, alive 
UC001 CML/AP NA 90 No II Ext 241 Progression 
UC002 CML/BC NA NA No NA NA 20 Progression 
FH15764 Hodgkin disease/rel 90 99 No III Ext > 581 Relapse, alive  
UL701 NHL/refr B cell 100* NA No IV NA 13 CR, VOD, ARDS  
SPN2219 NHL/rel diff. large cell 100 100 No Ext > 584 CR, alive 
FH13715 NHL/rel MCL 10 40 No No > 575 Relapse, alive 
FH16020 NHL/rel MCL 59 43 No II Ext > 548 CR, alive 
UL744 NHL/rel high-grade T cell NA No No 40 Progression 
FH15780 NHL/PR MCL 99 100 No II Ext > 451 CR, alive 
FH17028 WM/refr 81 90 No II Lim 293 Stable disease, infection 
FH15390 MDS/RA 60 Yes II NA 210 Relapse, infection 2nd BMT 
SPN2219 MDS/CMML 65 30 No No 178 Relapse 
SPN2264 MDS/RAEB-T NA NA No NA 18 NE, MOF, sepsis 
UL760 MDS/CMML 100 100 No No > 474 CR, alive 
FH11799 MDS/RAEB NA NA No II NA 21 NE, pneumonia 
FH15608 MDS/AML/CR Yes NA NA 215 Relapse 
FH16391 MDS/AML refr NA Yes NA NA 153 Relapse 
FH15595 MDS/MPS 43 65 No Ext > 470 Stable disease, alive 
UL664 MM/rel 100 100 No Lim 602 Relapse 
UL714 MM/rel 90 90 No No > 641 Progression, alive 
UL733 MM/rel 100 100 No II Lim 228 Progression 
UL747 MM/PR 98 100 No II Ext > 515 Relapse, alive 
UL753 MM/PR NA Yes NA NA 166 Stable disease, inf. 2nd BMT 
FH15711 MM/refr 90 NA No II NA 89 Progression, inf. 2nd BMT 
UL765 MM/PR 100 100 No IV NA 59 NE, aGVHD, bleeding 
FH14886 MM/refr 99 100 No II Ext 271 Progressive disease 
UL703 PNH 85 90 No II Lim > 683 CR, alive 
UPNDisease + status% Donor CD3 cellsGraft rejectionGVHDSurvival, dSummary of outcome as of 09/01/2001
Day 28Day 56Acute (grade)Chronic
SPN2292 ALL/CR2 90 100 No II Ext 211 CR, cGVHD, pneumonia/MOF 
UL755 ALL/CR2 83 88 No IV No 109 CR, aGVHD, infection 
SPN2292 AML/CR1 100 65 No II Lim > 549 CR, alive 
UL676 AML/CR2 85 100 No II Lim 156 CR, sudden death 
UL652 AML/CR3 100 100 No Lim > 844 CR, alive 
FH15303 AML/CR3 90 90 No III No 144 CR, GI bleed 
UL637 AML/CR3 100 100 No IV No 170 CR, aGVHD 
FH16411 AML/CR4 100 100 No II Ext 401 CR, CNS infection/sev. GvHD 
UL665 AML/PR2 75 80 No II Lim > 789 CR, alive 
UL668 AML/prog 96 99 No II Ext 624 CR, infection (urosepsis) 
UL680 AML/prog 95 99 No II Lim 218 Relapse 
UL692 AML/rel1 100 100 No II Lim > 705 CR, alive 
UL656 CLL/PR 100 100 No Lim > 817 CR, alive 
UL698 CLL/PR 88 86 No II Ext 482 CR, suicide, chronic GVHD 
FH16078 CLL/rel 99 99 No II Ext > 480 CR, alive 
UL597 CML/AP 75 80 No III No 122 Progression 
UL641 CML/AP 70 NA No II No 46 NE, TTP 
UL670 CML/AP NA Yes NA NA 172 Relapse 
UL672 CML/CP 75 85 No III Lim 546 CR, cGVHD 
UL695 CML/AP 100 100 No IV Lim > 698 CR, alive 
UL709 CML/CP 80 70 No IV Lim 312 CR, cGVHD 
UL725 CML/CP NA Yes NA NA 203 NE, infection 2nd BMT 
UL730 CML/CP 90 93 No IV No 61 NE, aGVHD 
UL735 CML/CP 95 100 No No 377 Relapse 
UL754 CML/CP2 90 100 No No > 492 CR, alive 
UC001 CML/AP NA 90 No II Ext 241 Progression 
UC002 CML/BC NA NA No NA NA 20 Progression 
FH15764 Hodgkin disease/rel 90 99 No III Ext > 581 Relapse, alive  
UL701 NHL/refr B cell 100* NA No IV NA 13 CR, VOD, ARDS  
SPN2219 NHL/rel diff. large cell 100 100 No Ext > 584 CR, alive 
FH13715 NHL/rel MCL 10 40 No No > 575 Relapse, alive 
FH16020 NHL/rel MCL 59 43 No II Ext > 548 CR, alive 
UL744 NHL/rel high-grade T cell NA No No 40 Progression 
FH15780 NHL/PR MCL 99 100 No II Ext > 451 CR, alive 
FH17028 WM/refr 81 90 No II Lim 293 Stable disease, infection 
FH15390 MDS/RA 60 Yes II NA 210 Relapse, infection 2nd BMT 
SPN2219 MDS/CMML 65 30 No No 178 Relapse 
SPN2264 MDS/RAEB-T NA NA No NA 18 NE, MOF, sepsis 
UL760 MDS/CMML 100 100 No No > 474 CR, alive 
FH11799 MDS/RAEB NA NA No II NA 21 NE, pneumonia 
FH15608 MDS/AML/CR Yes NA NA 215 Relapse 
FH16391 MDS/AML refr NA Yes NA NA 153 Relapse 
FH15595 MDS/MPS 43 65 No Ext > 470 Stable disease, alive 
UL664 MM/rel 100 100 No Lim 602 Relapse 
UL714 MM/rel 90 90 No No > 641 Progression, alive 
UL733 MM/rel 100 100 No II Lim 228 Progression 
UL747 MM/PR 98 100 No II Ext > 515 Relapse, alive 
UL753 MM/PR NA Yes NA NA 166 Stable disease, inf. 2nd BMT 
FH15711 MM/refr 90 NA No II NA 89 Progression, inf. 2nd BMT 
UL765 MM/PR 100 100 No IV NA 59 NE, aGVHD, bleeding 
FH14886 MM/refr 99 100 No II Ext 271 Progressive disease 
UL703 PNH 85 90 No II Lim > 683 CR, alive 
*

Day 12.

Death.

Infection after induction therapy for secondary AML.

NA indicates not available.

Of the 52 patients, 3 died too early to be evaluable for engraftment. Of the remaining 49 patients, 6 (12%; CI, 5%-25%) rejected their grafts between 21 and 56 days after HCT and had autologous marrow recoveries. Factors predictive of graft rejection in univariate analysis were low T-cell contents in the grafts (P = .04) and diagnosis of MDS (P = .03). Trends were observed for low numbers of transplanted CD34+cells (P = .06) and lack of preceding cytotoxic chemotherapy (P = .08). There were no correlations between rejection and HSC source (PBSC vs BM), patient or donor ages, and sex.

Regimen-related toxicities and infections

None of the patients experienced new-onset alopecia. Mild to moderate nausea due to MMF/CSP was common. Reversibly elevated serum creatinine or bilirubin levels ascribed to CSP were the most frequent toxicities (Table 3).

Table 3.

Maximum toxicities in 52 patients given unrelated HCT

GradeRenalHepaticCNSPulmonaryCardiacGIMucositisHemorrhagic
38 36 45 43 48 39 51 51 
II 
III 
IV 
GradeRenalHepaticCNSPulmonaryCardiacGIMucositisHemorrhagic
38 36 45 43 48 39 51 51 
II 
III 
IV 

Septicemias were encountered in 8 patients (15%; CI, 7%-28%). These were associated with pneumonia (n = 5), enteritis (n = 5), and/or sinusitis (n = 3). Three patients had toxoplasmosis, and one had pericarditis and pneumonia from Legionella pneumophila. One patient died of septicemia and one of pneumonia; all other infections were successfully treated with antibiotics. One patient died 624 days after transplantation after urosepsis and hydronephrosis caused by prostate hypertrophy.

Nine patients had proven or suspected pneumonias of fungal origin after HCT; 5 of these were successfully treated. Of the 4 patients with persisting pneumonia, one died of relapse, 2 died with GVHD, and one died after a second transplantation following failure of myeloid engraftment of the first.

HSV infections were observed in 6 patients (12%; 4%-23%), CMV reactivations/infections in 15 (29%; CI, 17%-43%), suspected viral pneumonitis in 4, varicella zoster virus (VZV) infection in 1, HHV6 infection in 1, Epstein-Barr virus (EBV) lymphoproliferative disease in 1, and suspected viral encephalitis in 3 patients. Lethal viral infections were not observed.

Acute GVHD

Thirty-three patients (63%; CI, 50%-77%) developed acute GVHD, which was grade II in 22 (42%;CI, 29%-56%), grade III in 4 (8%; CI, 0%-15%), and grade IV in 7 (13%, CI, 4%-23%; Table 2 and Figure3A). Acute GVHD was diagnosed at a median of 30 days (range, 8-284 days) after HCT and affected skin (n = 32), gut (n = 20), and liver (n = 10). Neither recipient and donor ages, recipient and donor sex, HSC source, underlying diseases, nor preceding treatment were predictive for acute GVHD. However, a correlation was suggested between HLA-class I disparity (P = .06) and grades III-IV acute GVHD.

Fig. 3.

Acute GVHD (A), relapse and transplantation-related mortality (B), and overall survival (C) after unrelated HCT.

Panel A shows the cumulative incidences of acute grades II, III, and IV GVHD among all 52 patients. Panel B shows the probabilities of disease progression and transplantation-related mortality for all 52 patients. Panel C shows survivals among high-risk and low-risk patients.

Fig. 3.

Acute GVHD (A), relapse and transplantation-related mortality (B), and overall survival (C) after unrelated HCT.

Panel A shows the cumulative incidences of acute grades II, III, and IV GVHD among all 52 patients. Panel B shows the probabilities of disease progression and transplantation-related mortality for all 52 patients. Panel C shows survivals among high-risk and low-risk patients.

Close modal

In most patients, GVHD responded to CSP and standard courses of methylprednisolone.24 In 5 patients, OKT3 was used because of steroid-resistant GVHD.

Incidence of chronic GVHD

Of 43 evaluable patients, 13 (30%) developed chronic GVHD requiring systemic immunosuppressive therapy.21 Eight patients died from complications associated with either acute or chronic GVHD (15%).

Status of underlying diseases

Table 2 summarizes the results.

Patients with graft rejection.

Of the 6 rejecting patients, 3 died from disease progression, and 3 died from infections after second HCT: one in complete remission (CR), one in partial remission (PR), and one with stable disease.

Patients with sustained engraftment.

Thirty-three patients had measurable disease before HCT, and 15 (45%) of these patients achieved CR at variable times after HCT. Two patients with ALL receiving transplants in second CR died in CR with infections with or without GVHD. Of 6 AML patients receiving transplants in first, third, or fourth CR, 2 were alive in CR, while 4 died in CR from transplantation complications. Of 4 AML patients with persistent disease who received transplants, 2 were alive in CR, one died following urosepsis in CR, and one died of relapse. Of 3 patients with CLL, 2 were alive in CR, while one died in CR from suicide. Of 10 patients with CML who received transplants in first or second chronic phase, accelerated phase, or blast crisis, 2 were alive in CR, 2 died in CR with complications from GVHD, 4 died from disease progression or relapse, and 2 died early (one from thrombotic thrombocytopenic purpura [TTP] and one with acute GVHD) without being evaluable for the underlying disease. Of 7 patients with NHL, 3 were in sustained CR after transplantation, 2 relapsed and are alive, one died of disease progression, and one died in CR of veno-occlusive disease/acute respiratory distress syndrome (VOD/ARDS). One patient with WM had stable disease, developed de novo AML of host cell origin, and died during induction therapy of his AML. Of 5 patients with MDS, 2 were alive, one in CR and one with stable disease, and 3 died: one with sepsis, one with pneumonia, and one with relapse. Of 7 patients with MM, 2 were alive in relapse, and 5 died, 4 either directly or indirectly from relapse and one with acute GVHD. The patient with PNH was alive and free of disease.

DLI

Six patients received DLI. One patient with AML achieved CR, relapsed, and did not respond to a second DLI. One patient with CML progressed to blast crisis after DLI and died. Of 4 patients with MM, 3 responded with transient PR, and one patient had no response.

Survival and causes of death

With a median follow-up of 19.3 months (range, 15.3-28.1 months), 18 of the 52 patients (35%) were alive. Of these, 13 were in CR, 1 had stable disease, and 4 have relapsed or progressed. Twelve patients (23%) died of causes related to transplantation, including GVHD (n = 8), pneumonia (n = 1), TTP (n = 1), VOD/ARDS (n = 1), and infection and multiorgan failure (n = 1). Among the remaining 22 patients who died, fourteen (27%) died from relapse/progression of their underlying diseases; one committed suicide on day 482 after HCT; one died from suspected myocardial infarction on day 156; one died from hemorrhage due to a congenital Dieulafoy arterial malformation25 on day 144; 3 died after rejection, disease recurrence, and second allogeneic HCT from infections; one died after induction therapy of de novo AML in host cells; and one died after urosepsis 624 days after transplantation. The median survival was 271 days.

Figures 3B and 3C show survival, progression, and transplantation-related mortality. Overall, survival was 44% (CI, 31%-58%) at 1 year. For low-risk patients, survival was 67% (CI, 29%-100%), and for high-risk patients, 41 (CI, 27%-56%). There was no statistically significant difference in survival between HLA-matched and mismatched patients (Tables 1 and 2). Overall mortality directly related to the first unrelated donor transplantation was 29 (CI, 17%-41%) at one year. Mortality from disease progression was 27% (CI, 15%-39%) at one year. In a multivariate analysis, rejection (P = .03) and HLA class II mismatch (P = .04) adversely affected survival. Not surprisingly, rejection was a risk factor for relapse (P = .02).

This study extends previous observations made in HLA-identical siblings to include grafts from HLA-matched or mismatched unrelated donors. None of the patients were eligible for conventional HCT, either because of age (median age, 48 years) or medical contraindications. Most (88%) were considered high-risk patients, and 42% had failed previous autologous or allogeneic HCT. The study results allowed several conclusions.

First, the regimen was well tolerated and minimally toxic without the typical side effects of conventional HCT. Of 3 patients who died within the first 3 weeks, 2 died from pre-existing infections, which had made them ineligible for conventional HCT, and the third had a long history of chemotherapy, radiation therapy, and 2 unsuccessful high-dose conventional autologous HCTs in addition to developing hyperacute GVHD. The day-100 transplantation-related mortality was 11%. This compared favorably to the 29% day-100 mortality among younger (median age, 20 years) patients with acute leukemias given conventional unrelated HCT26 and to the 41% day-100 mortality among younger patients (median age, 28 years) given conventional allogeneic HCT after preceding failed autologous HCT.27 In addition, even though many patients had abnormally low granulocyte and platelet counts before transplantation, severe protracted pancytopenias were generally not encountered after transplantation. Because of the mild myelosuppression of the current regimen, many patients were managed either as outpatients or in regular hospital rooms and generally did not require hematopoietic growth factor support as has been reported by others.9,12 Transfusion needs were significantly lower than those among patients given conventional HCT.28Considering the patients' ages, the extent of preceding therapies, and their pre-existing organ dysfunction or infections, the regimen had acceptable toxicities. The observed 100-day transplantation-related mortality of 11% compared favorably to those in another recent study, which ranged from 37.4%-87.5%, depending on the reduced-intensity conditioning regimen used and on whether hematopoietic cell donors were related or unrelated.9 In a second study of much younger patients (median age, 17 years [range, 8-48 years] ) undergoing reduced-intensity unrelated HCT, 2 (12.5%) of 16 patients died of transplantation-related complications within 100 days of HCT.10 A third recent study of 47 patients undergoing unrelated HCT showed a day-100 mortality of 14.9%. In this study, patient ages were lower (median, 44 years; range, 18-62 years) than in the current study, and patients with a life expectancy of < 8 weeks were excluded.12 

Second, sustained allogeneic engraftment was achieved in 88% of evaluable patients, and graft rejections (n = 6) did not lead to fatal pancytopenias. Rejections occurred predominantly among patients with MDS and those with low T-cell contents in their grafts. They were characterized by either no appearance of donor cells (n = 5) or initial appearance and subsequent gradual disappearance of such cells within 56 days of HCT (n = 1). In all rejecting patients, the underlying diseases persisted and progressed, leading either directly or indirectly (failed second HCT) to death. With the limited data available, there was no discernable correlation between HLA-mismatching and rejection. We previously reported a 5% graft rejection rate among patients (median age, 35 years) with CML in chronic phase (CP) given conventional unrelated HCT,1 a rate that would appear lower than the one reported here (12%), although others have described rejection rates of 15.5% for CML patients3 and of 13% for ALL patients29 after conventional HCT, though the relatively high rate of rejection among CML patients could be due to the use of T-cell–depleted grafts among some of the patients. Others carried out unrelated grafts after reduced intensity conditioning regimens and reported either no graft rejection10 or provided no detailed information.9 Among current patients with engraftment, conversion from host to all-donor hematopoietic cells was generally complete between 3 and 6 months after HCT. Ways to avoid graft rejection in future protocols include the use of grafts with high T-cell contents and modified protocols that convey a higher degree of pretransplantation immunosuppression to overcome transfusion-induced sensitization in patients with MDS who often had no preceding chemotherapies. Optimizing MMF therapy using level-adapted dosing might be another promising approach.

Third, 42% of evaluable patients developed grade II; 8%, grade III; and 13%, grade IV acute GVHD. There was a suggested correlation between GVHD and HLA class I disparity. The current 63% rate of GVHD was comparable to rates previously reported in younger patients with acute leukemias (82%) and CML in CP (77%-89%) given conventional HCT1,26; however, the current 21% rate of grades III-IV GVHD appeared lower than the previously reported rates of 35%-47%. None of the patients with grade II and III acute GVHD died from this complication; 9% of evaluable patients died with grade IV acute GVHD. While this value appeared lower than previously reported after conventional HCT, improved GVHD control remains a critical future research objective. Chronic GVHD requiring therapy developed in 30% of patients with durable engraftment. The relatively benign course of GVHD among these elderly patients with unrelated HLA-matched and -mismatched HCT compared to conventional HCT could be the result of potent immunosuppression by the combination of MMF/CSP, although contributions of the low-intensity pretransplantation conditioning, for example, lack of tissue injury and “cytokine storm”30 and the initial mixed donor-host chimerism, also might be important. A study of unrelated HCT after a conditioning regimen containing a purine analog and melphalan reported 23% grade II and 39% grade III-IV acute GVHD, and deaths related to GVHD were observed in 27.5% of patients.9 Another study using a purine analog and melphalan in combination with CAMPATH-1H monoclonal antibody conditioning reported 14.9% grade II and 6.4% grade III-IV acute GVHD and 8% limited chronic GVHD.12 A comparison of current results with unrelated donors to those previously reported with related donors given the same regimen11 shows higher incidences of acute GVHD (63% vs 47%) and nonrelapse mortality (29% vs 6.7%). Further reduction in the incidence of GVHD might be achieved in the future by optimizing the duration of immunosuppressive therapy after transplantation and by using HLA allele–matched donors.

Fourth, no clear relationship emerged between HLA antigen or allele level mismatching and graft rejection, though a suggested correlation between HLA class I mismatch and GVHD was found. Also, HLA class II mismatch adversely affected survival.

Finally, graft-versus-tumor responses occurred among patients with sustained grafts. Of patients with measurable disease before HCT, 45% attained CR, including 6 of 16 (38%) evaluable patients with preceding failed high-dose autologous or allogeneic HCT. It is unlikely that low-dose TBI and fludarabine were responsible for the CR given that those patients, for the most part, had failed to respond to more specific and intensive therapies, including the high-dose regimens used for HCT. Thus, the current findings are most consistent with graft-versus-tumor effects. These were seen early after transplantation, especially in patients with acute myeloid leukemia, where patients in persistent disease reached remissions within a few weeks after transplantation. NK cells with killer cell inhibitory receptor (KIR) mismatches might have played a role in these rapid-onset graft-versus-tumor reactions, which were usually not associated with GVHD.31 Correlations of KIR mismatches with clinical outcome will be analyzed in current donor-patient combinations. In other diseases such as CML or CLL, graft-versus-tumor effects required months to induce remissions and became evident often only after discontinuation of immunosuppression. Selective tumor cell killing induced by tumor-specific T cells might have played a role in these remissions, and this is currently under study. Relapse mortality was comparable in the present study of unrelated HCT (25%) and in the previously published study of related HCT (27%),11 though there was a greater number of high-risk patients in the current study. The role of DLIs in current transplantations is still unclear. Only a minority of patients received DLIs, and all of them had only transient responses. In the future, more selective tumor cell killing may be accomplished through focusing T-cell immunity against tumor antigens or minor histocompatibility antigens with hematopoietic restriction.32-34 

In conclusion, this phase 1 study shows that fludarabine and 2 Gy TBI in combination with cyclosporine and MMF are sufficient to obtain engraftment in most patients with unrelated HLA-matched and partially mismatched donors. Phase 2 studies will evaluate the usefulness of the graft-versus-tumor effect for individual hematological malignancies.

We thank the medical, nursing, laboratory, data processing, and secretarial staff of the participating institutions for their important contributions to this study, including the careful and dedicated care of the patients.

Prepublished online as Blood First Edition Paper,October 3, 2002; DOI 10.1182/blood-2002-05-1340.

Supported in part by grants CA78902, CA49605, HL36444, CA18221, CA18029, CA15704, AI49213, and HL03701 awarded by the National Institutes of Health, Department of Health and Human Services, Bethesda, MD; a grant from the Gabrielle Rich Leukemia Foundation (P.A.M., D.G.M.); and support from the Laura Landro Salomon Endowment Fund (F.R.S.). Chimerism studies in patients treated at University of Leipzig were supported by a grant from Amgen, Lucerne, Switzerland.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Hansen
JA
Gooley
TA
Martin
PJ
et al
Bone marrow transplants from unrelated donors for patients with chronic myeloid leukemia.
N Engl J Med.
338
1998
962
968
2
Kernan
NA
Bartsch
G
Ash
RC
et al
Analysis of 462 transplantations from unrelated donors facilitated by The National Marrow Donor Program.
N Engl J Med.
328
1993
593
602
3
McGlave
PB
Shu
XO
Wen
W
et al
Unrelated donor marrow transplantation for chronic myelogenous leukemia: 9 years' experience of the National Marrow Donor Program.
Blood.
95
2000
2219
2225
4
Molina
AJ
Storb
RF
Hematopoietic stem cell transplantation in older adults.
Handbook of Bone Marrow Transplantation.
1st ed.
Rowe
JM
Lazarus
HM
Carella
AM
2000
111
137
Martin Dunitz
London, United Kingdom
5
Slavin
S
Nagler
A
Naparstek
E
et al
Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases.
Blood.
91
1998
756
763
6
Khouri
IF
Keating
M
Körbling
M
et al
Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies.
J Clin Oncol.
16
1998
2817
2824
7
Childs
R
Clave
E
Contentin
N
et al
Engraftment kinetics after nonmyeloablative allogeneic peripheral blood stem cell transplantation: full donor T-cell chimerism precedes alloimmune responses.
Blood.
94
1999
3234
3241
8
Sykes
M
Preffer
F
McAfee
S
et al
Mixed lymphohaemopoietic chimerism and graft-versus-lymphoma effects after non-myeloablative therapy and HLA-mismatched bone-marrow transplantation.
Lancet.
353
1999
1755
1759
9
Giralt
S
Thall
PF
Khouri
I
et al
Melphalan and purine analog-containing preparative regimens: reduced-intensity conditioning for patients with hematologic malignancies undergoing allogeneic progenitor cell transplantation.
Blood.
97
2001
631
637
10
Nagler
A
Aker
M
Or
R
et al
Low-intensity conditioning is sufficient to ensure engraftment in matched unrelated bone marrow transplantation.
Exp Hematol.
29
2001
362
370
11
McSweeney
PA
Niederwieser
D
Shizuru
JA
et al
Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects.
Blood.
97
2001
3390
3400
12
Chakraverty
R
Peggs
K
Chopra
R
et al
Limiting transplantation-related mortality following unrelated donor stem cell transplantation by using a nonmyeloablative conditioning regimen.
Blood.
99
2002
1071
1078
13
Storb
R
Yu
C
Wagner
JL
Deeg
HJ
et al
Stable mixed hematopoietic chimerism in DLA-identical littermate dogs given sublethal total body irradiation before and pharmacological immunosuppression after marrow transplantation.
Blood.
89
1997
3048
3054
14
Petersdorf
EW
Gooley
TA
Anasetti
C
et al
Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient.
Blood.
92
1998
3515
3520
15
Petersdorf
EW
Gooley
T
Malkki
M
et al
The biological significance of HLA-DP gene variation in haematopoietic cell transplantation.
Br J Haematol.
112
2001
988
994
16
Meyers
JD
Petersen
FB
Counts
GW
et al
Bacterial, fungal and protozoan infection after marrow transplantation.
Recent Advances and Future Directions in Bone Marrow Transplantation: Experimental Hematology Today—1987.
Baum
SJ
Santos
GW
Takaku
F
1988
171
176
Springer Verlag
New York, NY
17
Slavin
MA
Osborne
B
Adama
R
et al
Efficacy and safety of fluconazole prophylaxis for fungal infections after marrow transplantation: a prospective, randomized, double-blind study.
J Infect Dis.
171
1995
1545
1552
18
Boeckh
M
Bowden
RA
Goodrich
JM
Pettinger
M
Meyers
JD
Cytomegalovirus antigen detection in peripheral blood leukocytes after allogeneic marrow transplantation.
Blood.
80
1992
1358
1364
19
Bryant
E
Martin
PJ
Documentation of engraftment and characterization of chimerism following hematopoietic cell transplantation.
Hematopoietic Cell Transplantation.
2nd ed.
Thomas
ED
Blume
KG
Forman
SJ
1999
197
206
Blackwell Science
Boston, MA
20
Przepiorka
D
Weisdorf
D
Martin
P
et al
Consensus conference on acute GVHD grading.
Bone Marrow Transplant.
15
1995
825
828
21
Sullivan
KM
Agura
E
Anasetti
C
et al
Chronic graft-versus-host disease and other late complications of bone marrow transplantation.
Semin Hematol.
28
1991
250
259
22
Radich
J
Ladne
P
Gooley
T
Polymerase chain reaction-based detection of minimal residual disease in acute lymphoblastic leukemia predicts relapse after allogeneic BMT.
Biol Blood Marrow Transplant.
1
1995
24
31
23
Andersen
PK
Borgan
O
Gill
RD
Keiding
N
Statistical Models Based on Counting Processes.
1993
Springer-Verlag
New York, NY
24
Martin
PJ
Schoch
G
Fisher
L
et al
A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment.
Blood.
76
1990
1464
1472
25
Blecker
D
Bansal
M
Zimmerman
RL
et al
Dieulafoy's lesion of the small bowel causing massive gastrointestinal bleeding: two case reports and literature review (review).
Am J Gastroenterol.
96
2001
902
905
26
Sierra
J
Storer
B
Hansen
JA
et al
Transplantation of marrow cells from unrelated donors for treatment of high-risk acute leukemia: the effect of leukemic burden, donor HLA-matching, and marrow cell dose.
Blood.
89
1997
4226
4235
27
Radich
JP
Gooley
T
Sanders
JE
Anasetti
C
Chauncey
T
Appelbaum
FR
Second allogeneic transplantation after failure of first autologous transplantation.
Biol Blood Marrow Transplant.
6
2000
272
279
28
Weissinger
F
Sandmaier
BM
Maloney
DG
Bensinger
WI
Gooley
T
Storb
R
Decreased transfusion requirements for patients receiving nonmyeloablative compared with conventional peripheral blood stem cell transplants from HLA-identical siblings.
Blood.
98
2001
3584
3588
29
Cornelissen
JJ
Carston
M
Kollman
C
et al
Unrelated marrow transplantation for adult patients with poor-risk acute lymphoblastic leukemia: strong graft-versus-leukemia effect and risk factors determining outcome.
Blood.
97
2001
1572
1577
30
Ferrara
JL
Pathogenesis of acute graft-versus-host disease: cytokines and cellular effectors (review).
J Hematother Stem Cell Res.
9
2000
299
306
31
Ruggeri
L
Capanni
M
Urbani
E
et al
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants.
Science.
295
2002
2097
2100
32
Goulmy
E
Human minor histocompatibility antigens: new concepts for marrow transplantation and adoptive immunotherapy (review).
Immunol Rev.
157
1997
125
140
33
Warren
EH
Gavin
MA
Simpson
E
et al
The human UTY gene encodes a novel HLA-B8-restricted H-Y antigen.
J Immunol.
164
2000
2807
2814
34
Mutis
T
Verdijk
R
Schrama
E
Esendam
B
Brand
A
Goulmy
E
Feasibility of immunotherapy of relapsed leukemia with ex vivo-generated cytotoxic T lymphocytes specific for hematopoietic system-restricted minor histocompatibility antigens.
Blood.
93
1999
2336
2341

Author notes

Dietger Niederwieser, Division of Hematology and Oncology, University of Leipzig, Philipp Rosenthalstr 23-25, D-4103 Leipzig, Germany; e-mail:dietger@medizin.uni-leipzig.de.

Sign in via your Institution