Toxicities have limited the use of allogeneic hematopoietic cell transplantation (HCT) to younger, medically fit patients. In a canine HCT model, a combination of postgrafting mycophenolate mofetil (MMF) and cyclosporine (CSP) allowed stable allogeneic engraftment after minimally toxic conditioning with low-dose (200 cGy) total-body irradiation (TBI). These findings, together with the known antitumor effects of donor leukocyte infusions (DLIs), led to the design of this trial. Forty-five patients (median age 56 years) with hematologic malignancies, HLA-identical sibling donors, and relative contraindications to conventional HCT were treated. Immunosuppression involved TBI of 200 cGy before and CSP/MMF after HCT. DLIs were given after HCT for persistent malignancy, mixed chimerism, or both. Regimen toxicities and myelosuppression were mild, allowing 53% of eligible patients to have entirely outpatient transplantations. Nonfatal graft rejection occurred in 20% of patients. Grades II to III acute graft-versus-host disease (GVHD) occurred in 47% of patients with sustained engraftment. With median follow-up of 417 days, survival was 66.7%, nonrelapse mortality 6.7%, and relapse mortality 26.7%. Fifty-three percent of patients with sustained engraftment were in complete remission, including 8 with molecular remissions. This novel allografting approach, based on the use of postgrafting immunosuppression to control graft rejection and GVHD, has dramatically reduced the acute toxicities of allografting. HCT with the induction of potent graft-versus-tumor effects can be performed in previously ineligible patients, largely in an outpatient setting. Future protocol modifications should reduce rejection and GVHD, thereby facilitating studies of allogeneic immunotherapy for a variety of malignancies.

Allogeneic hematopoietic cell transplantation (HCT) is effective treatment for patients with hematologic malignancies. Current strategies rely on maximally tolerated doses of systemic chemoradiation to eradicate cancer, and the allografts serve to rescue patients from treatment-induced marrow aplasia. Associated toxicities have limited treatment to younger, medically fit patients, with therapy administered on specialized hospital wards. Almost no HCTs have been done in patients more than 60 years of age, and relatively few in patients older than 50 years.1 Consequently, median patient ages at HCT for chronic myelocytic leukemia (CML), chronic lymphocytic leukemia (CLL), acute myelocytic leukemia (AML), multiple myeloma (MM), and non-Hodgkin lymphoma (NHL) are approximately 2 decades younger than median ages at diagnoses.1 Thus, current allogeneic HCT benefits only a younger minority of patients with candidate diseases.

At least 2 observations have raised doubts as to whether current concepts of allogeneic HCT are universally valid. First, marrow-based malignancies cannot always be eradicated even by the most intensive pretransplantation therapies.2,3 Second, cures may be attributed to graft-versus-tumor (GVT) reactions mediated by donor T cells,4-10 and donor lymphocyte infusions (DLIs) can induce sustained complete remissions (CRs) of malignancies that relapsed after HCT.11-16 These observations, together with a better understanding of how to influence immune cell functions, suggested a reevaluation of how to perform HCT. Specifically, the emphasis is shifting away from relying on high-dose cytotoxic therapy toward using donors' immune cells for eradicating malignancy. Consequently, some investigators have tested reduced-intensity pretransplantation regimens, although these regimens were still given for tumor cytoreduction and had significant toxicities, including temporary severe myelosuppression.17-20 

Here, we present a novel concept of allogeneic HCT that uses the knowledge that both host-versus-graft (HVG) and graft-versus-host (GVH) reactions are mediated by T cells. The concept relies on optimizing pharmacologic postgrafting immunosuppression to control both reactions. In canine HCT studies using major histocompatibility complex-matched littermate donors, the synergistic combination of the antimetabolite mycophenolate mofetil (MMF) and the T-cell–activation blocker cyclosporine (CSP) prevented graft-versus-host disease (GVHD) and reduced HVG reactions, thereby allowing a reduction of the single-dose pretransplantation total-body irradiation (TBI) needed for sustained engraftment from 920 cGy to a nonmyeloablative dose of 200 cGy.21-23 Dogs having transplantation this way experienced minimal toxicities and mild pancytopenia, and moreover became stable mixed hematopoietic chimeras. These observations, together with the known GVT effects of allografts, allowed us to apply a novel strategy of HLA-identical sibling HCT using low-dose (200 cGy) TBI/MMF/CSP. In contrast, conventional allografting regimens typically use TBI doses 6 times higher (at least 1200 cGy) combined with high-dose chemotherapy (eg, cyclophosphamide 120 mg/kg). This report describes initial results in 45 patients with hematologic malignancies who were ineligible for conventional HCT because of age or medical contraindications.

Patients and eligibility

Forty-five consecutive patients were treated between December 17, 1997, and May 27, 1999, at the Fred Hutchinson Cancer Research Center (FH), University of Washington Medical Center, and Veterans Affairs Medical Center in Seattle; the University of Leipzig (UL); and Stanford University (SU). Consent was obtained according to each institution's institutional review board guidelines.

Major eligibility requirements were as follows: (1) indolent hematologic malignancy or acute leukemia in CR; (2) HLA-identical sibling donor with serologic match at HLA-A, -B, -C, and molecular matching for HLA-DRB124; (3) age greater than 50 years (greater than 60 years for CML) or relative contraindication to conventional HCT in younger patients; and (4) creatinine clearance greater than 50 mL/min, bilirubin less than 2 times normal, and Karnofsky score more than 50. CLL and lymphoma patients had failed at least frontline therapy. MM patients had stage II or III disease at diagnosis or disease progression and had had previous chemotherapy. Three additional poor-risk patients had chemotherapy-resistant acute leukemia (UL607, UL577) and persistent marrow aplasia and aspergillosis after induction chemotherapy for AML (FH14241).25 Table1 shows patient and graft characteristics.

Table 1.

Characteristics of patients and grafts (grouped by rejection status and diagnoses)

Unique patient no.DiagnosisAge/sex*Previous autograftPrevious regimensCD34+ cells × 106/kgCD3+ cells × 108/kg1-153
UL 607 ALL-REL3 60/M  VDAspAra, IdaAra-C, FLAG × 2, FLAG × 2 3.9 4.5 
 577 MF-AML-IF 62/M  Hu, IFN, IdaTG × 6, IdaAra-C × 2, E Ida (all for MF), HDAC, Mi 4.8 4.3  
FH 14241 AML-aplasia 49/M  IdaAra-C × 2 → 10 weeks of aplasia + aspergillosis 10.7 2.7 
 14560 AML-CR1 63/M  IdaAra-C × 2, HDAC 6.5 2.4 
 13461 AML-CR1 60/M  MCE × 2, AraC 6.1 6.7 
UL 646 AML-CR1 36/F  IdaAra-C × 3 4.6 8.3 
 627 AML-CR1 58/F  IdaAra-C × 3 9.1 4.2 
 636 AML-CR1 56/F  IdaAra-C × 3 8.0 4.5  
UL 616 AML-CR2 72/M  TAD, HAM, Mi-FLAG 3.1 2.4  
FH 13361 AML-CR3 66/M  D Ara-C × 3, D Ara-C × 2, IdaAra-C × 2 4.0 0.7  
SU 1857 AML-IF 57/M  CMOPP/CHOP, XRT (for NHL), IdaAra-C for secondary AML 6.9 3.1  
FH 12914 CLL-REL2 53/M  F × 6, F × 8 11.3 4.9  
SU 1879 CLL-PR2 59/M  FChP × 8, F × 8 5.5 2.0  
FH 14587 CLL-REF 54/M  F × 6, CVP × 2 5.2 1.9 
 14586 CLL-REF 53/M  F × 6 9.6 3.0  
SU 2028 CLL-PR2 55/M  CHOP × 6, CyE + Rituxin × 4 5.1 2.6  
FH 14322 CLL-REF 46/M  Ch, CVP × 4, F × 4, CED 10.8 3.9  
 14627 CLL-REF 54/F  F × 6, CHOP × 4, CVP × 4 5.0 3.4  
UL 596 CML-AP 64/M  Hu/IFN, Hu/TG 4.7 4.3  
UL 605 CML-CP 49/F  Hu/IFN, TG 10.2 2.9  
FH 14214 CML-CP 70/M  Hu 3.3 1.7  
UL 634 CML-CP 60/M  Hu, IdaAra-C, Hu/IFN 5.9 2.5  
FH 14726 CML-CP 61/F  Hu, IFN 6.4 4.1  
SU 1955 HD-CR2 61/F  ABVD × 6, CHAD × 3 8.6 4.6  
FH 13817 HD-PR 49/F  XRT, MOPP × 3, ABVD × 6, DHAP × 3, MOPP × 2 16.1 4.8 
 14226 HD-REF 31/M Yes ABVD × 6, mantle XRT, MOPP × 3, HDT, Vb, GcCp × 2, Gc 12.3 3.6 
 14692 HD-REF 39/M Yes MOPP/ABVD × 6, ABVD × 5, DHAP × 2, HDT 4.7 2.4  
 13527 MM-PR 56/M Yes MP, VAD × 3, CyTx, HDT 3.3 2.6 
 13922 MM-PR 58/F Yes VAD × 4, CyTx, HDT 7.2 0.8  
 11924 MM-PR 44/F Yes VAD × 4, CyTx, HDT 17.5 5.2  
 13541 MM-REF 55/F Yes VAD × 4, CyTx, HDT 12.4 4.0  
 14274 MM-REF 63/M Yes VAD × 7, CyTx, HDT 15.5 6.0  
UL 625 MM-REF 59/M  MP, Brendamustine + P 9.7 2.9  
FH 14224 NHL-REF 53/F  Ch, FMD × 3, Promace-Cytabom × 4, Rituxin × 4, CE × 2 6.0 7.2 
 14413 WM-REF 50/M  F × 7, Rituxin × 4, Dex × 10 10.5 2.6  
 14658 WM-REF 58/M  2-CDA × 2, CVP × 2 13.2 4.9  
UL 644 AML-CR1 52/F  IdaAra-C × 3 3.8 3.5  
 14284 CLL-PR 67/M  Ch, F × 3 6.5 4.3  
 14091 CML-AP 54/M  Hu/INF, 2-CDA 15.1 2.0  
SU 2000 CML-AP 40/M  Hu 7.7 4.5  
SU 1948 CML-CP 71/M  Hu/INF, Ara-C/Hh, Ara-C 12.7 3.3 
UL 622 CML-CP 58/M  Hu 10.8 3.7  
FH 13009 MDS-RAEB-T 43/M  None 4.4 2.7 
 13246 MM-REF 58/F  VAD × 4 9.1 1.0 
 13230 MM-PR 53/M  VAD × 7 10.0 3.3 
Unique patient no.DiagnosisAge/sex*Previous autograftPrevious regimensCD34+ cells × 106/kgCD3+ cells × 108/kg1-153
UL 607 ALL-REL3 60/M  VDAspAra, IdaAra-C, FLAG × 2, FLAG × 2 3.9 4.5 
 577 MF-AML-IF 62/M  Hu, IFN, IdaTG × 6, IdaAra-C × 2, E Ida (all for MF), HDAC, Mi 4.8 4.3  
FH 14241 AML-aplasia 49/M  IdaAra-C × 2 → 10 weeks of aplasia + aspergillosis 10.7 2.7 
 14560 AML-CR1 63/M  IdaAra-C × 2, HDAC 6.5 2.4 
 13461 AML-CR1 60/M  MCE × 2, AraC 6.1 6.7 
UL 646 AML-CR1 36/F  IdaAra-C × 3 4.6 8.3 
 627 AML-CR1 58/F  IdaAra-C × 3 9.1 4.2 
 636 AML-CR1 56/F  IdaAra-C × 3 8.0 4.5  
UL 616 AML-CR2 72/M  TAD, HAM, Mi-FLAG 3.1 2.4  
FH 13361 AML-CR3 66/M  D Ara-C × 3, D Ara-C × 2, IdaAra-C × 2 4.0 0.7  
SU 1857 AML-IF 57/M  CMOPP/CHOP, XRT (for NHL), IdaAra-C for secondary AML 6.9 3.1  
FH 12914 CLL-REL2 53/M  F × 6, F × 8 11.3 4.9  
SU 1879 CLL-PR2 59/M  FChP × 8, F × 8 5.5 2.0  
FH 14587 CLL-REF 54/M  F × 6, CVP × 2 5.2 1.9 
 14586 CLL-REF 53/M  F × 6 9.6 3.0  
SU 2028 CLL-PR2 55/M  CHOP × 6, CyE + Rituxin × 4 5.1 2.6  
FH 14322 CLL-REF 46/M  Ch, CVP × 4, F × 4, CED 10.8 3.9  
 14627 CLL-REF 54/F  F × 6, CHOP × 4, CVP × 4 5.0 3.4  
UL 596 CML-AP 64/M  Hu/IFN, Hu/TG 4.7 4.3  
UL 605 CML-CP 49/F  Hu/IFN, TG 10.2 2.9  
FH 14214 CML-CP 70/M  Hu 3.3 1.7  
UL 634 CML-CP 60/M  Hu, IdaAra-C, Hu/IFN 5.9 2.5  
FH 14726 CML-CP 61/F  Hu, IFN 6.4 4.1  
SU 1955 HD-CR2 61/F  ABVD × 6, CHAD × 3 8.6 4.6  
FH 13817 HD-PR 49/F  XRT, MOPP × 3, ABVD × 6, DHAP × 3, MOPP × 2 16.1 4.8 
 14226 HD-REF 31/M Yes ABVD × 6, mantle XRT, MOPP × 3, HDT, Vb, GcCp × 2, Gc 12.3 3.6 
 14692 HD-REF 39/M Yes MOPP/ABVD × 6, ABVD × 5, DHAP × 2, HDT 4.7 2.4  
 13527 MM-PR 56/M Yes MP, VAD × 3, CyTx, HDT 3.3 2.6 
 13922 MM-PR 58/F Yes VAD × 4, CyTx, HDT 7.2 0.8  
 11924 MM-PR 44/F Yes VAD × 4, CyTx, HDT 17.5 5.2  
 13541 MM-REF 55/F Yes VAD × 4, CyTx, HDT 12.4 4.0  
 14274 MM-REF 63/M Yes VAD × 7, CyTx, HDT 15.5 6.0  
UL 625 MM-REF 59/M  MP, Brendamustine + P 9.7 2.9  
FH 14224 NHL-REF 53/F  Ch, FMD × 3, Promace-Cytabom × 4, Rituxin × 4, CE × 2 6.0 7.2 
 14413 WM-REF 50/M  F × 7, Rituxin × 4, Dex × 10 10.5 2.6  
 14658 WM-REF 58/M  2-CDA × 2, CVP × 2 13.2 4.9  
UL 644 AML-CR1 52/F  IdaAra-C × 3 3.8 3.5  
 14284 CLL-PR 67/M  Ch, F × 3 6.5 4.3  
 14091 CML-AP 54/M  Hu/INF, 2-CDA 15.1 2.0  
SU 2000 CML-AP 40/M  Hu 7.7 4.5  
SU 1948 CML-CP 71/M  Hu/INF, Ara-C/Hh, Ara-C 12.7 3.3 
UL 622 CML-CP 58/M  Hu 10.8 3.7  
FH 13009 MDS-RAEB-T 43/M  None 4.4 2.7 
 13246 MM-REF 58/F  VAD × 4 9.1 1.0 
 13230 MM-PR 53/M  VAD × 7 10.0 3.3 

UL indicates University of Leipzig; ALL, acute lymphocytic leukemia; REL, untreated relapse; MF, myelofibrosis; AML, acute myelogenous leukemia; IF, chemotherapy induction failure; FH, Fred Hutchinson Cancer Research Center; CR, complete remission; SU, Stanford University; CLL, chronic lymphocytic leukemia; PR, partial remission; REF, refractory to conventional chemotherapy; CML, chronic myelogenous leukemia; AP, accelerated phase; CP, chronic phase; HD, Hodgkin disease; MM, multiple myeloma; NHL, non-Hodgkin lymphoma; WM, Waldenström macroglobulinemia; MDS, myelodysplastic syndrome; RAEB-T, refractory anemia with excess blasts in transformation.

*

Median age = 56 years.

Previous regimens: eg, ×2 = 2 cycles. Abbreviations are expanded in the at the end this article.

Median CD34+ cell dose = 9 × 106/kg.

F1-153

Median CD3+ cell dose = 3.3 × 108/kg.

Treatment and evaluations

Figure 1 shows the treatment protocol. TBI was given at 7 cGy/min from dual cobalt 60 sources (Fred Hutchinson Cancer Research Center) or linear accelerators (Veterans Affairs Medical Center, University of Leipzig, Stanford University, University of Washington Medical Center). Forty-four patients received 2 Gy TBI alone. One patient (FH14726) with CML received, in addition, fludarabine 30 mg/m2 on days −4, −3, and −2 in an attempt to reduce the risk of graft rejection. CSP levels were targeted to the upper therapeutic range (approximately 500 ng/mL; Abbott TDX, Abbott Park, IL) until day +35. Prophylaxis against Pneumocystis carinii, fungal infections, and cytomegalovirus (CMV) infections was used.26-28 Patients with chronic GVHD requiring systemic immunosuppression received antibiotic prophylaxis againstP carinii and Pneumococcus. For outpatient transplantations, scheduled follow-up included 2 to 3 clinic visits per week for the first month and then once or twice per week or as clinically required thereafter.

Fig. 1.

Treatment protocol for nonmyeloablative HCT.

Granulocyte colony-stimulating factor (G-CSF)–mobilized peripheral blood stem cells (PBSCs) were infused after TBI on day 0. One patient (FH14726) received fludarabine 30 mg/m2 ×3 intravenously on days −4, −3, and −2 before TBI/MMF/CSP. G-CSF: 16 μg per kg per day on days −4 to 0, aphereses on days −1, 0; TBI: 200 cGy (7 cGy/min) single fraction; HCT: PBSCs infused on day 0; CSP: 1.5 mg/kg intravenously twice daily on days −1 and 0, 6.25 mg/kg orally twice daily on days 1 to +35 (cohort 1), then taper to +56 (cohort 2); MMF: 15 mg/kg orally twice daily on days 0 to +27; DLI: no. 1 equals 107 CD3+ cells/kg, no. 2 equals 3.3 × 107 CD3+ cells/kg.

Fig. 1.

Treatment protocol for nonmyeloablative HCT.

Granulocyte colony-stimulating factor (G-CSF)–mobilized peripheral blood stem cells (PBSCs) were infused after TBI on day 0. One patient (FH14726) received fludarabine 30 mg/m2 ×3 intravenously on days −4, −3, and −2 before TBI/MMF/CSP. G-CSF: 16 μg per kg per day on days −4 to 0, aphereses on days −1, 0; TBI: 200 cGy (7 cGy/min) single fraction; HCT: PBSCs infused on day 0; CSP: 1.5 mg/kg intravenously twice daily on days −1 and 0, 6.25 mg/kg orally twice daily on days 1 to +35 (cohort 1), then taper to +56 (cohort 2); MMF: 15 mg/kg orally twice daily on days 0 to +27; DLI: no. 1 equals 107 CD3+ cells/kg, no. 2 equals 3.3 × 107 CD3+ cells/kg.

Close modal

Chimerism among peripheral blood (PB) T cells, granulocytes, and unfractionated marrow was 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 sex-mismatched transplants and polymerase chain reaction (PCR)–based analysis of polymorphic microsatellite regions for recipients of sex-matched transplants.29 

The primary study end point was mixed chimerism at day 28, defined as between 1% and 95% PB donor T cells. Patients with mixed chimerism, no GVHD, and less than 20% increases in donor T cells from day 28 to 56 were eligible for DLI on day +65. In others, monthly chimerism and tumor evaluations were done to assess the need for DLI. DLIs were scheduled to be administered at 2-month intervals starting at 1 × 107 CD3+ cells/kg, with up to 3 subsequent infusions at doses of 3.3 × 107, 1.0 × 108, and 3.3 × 108 CD3+cells/kg to be given based on the presence of stable mixed chimerism and absence of GVHD, or persistent or progressive disease. Patients who had rapidly progressive tumors early after transplantation or graft rejection were considered treatment failures and were eligible for other therapies including DLI off protocol or second transplantations.

Acute and chronic GVHD were graded as described.30,31Tumor responses were assessed using standard criteria. In patients who achieved CR, PCR was used to detect bcr-abltranscripts (CML)32 or clonal immunoglobulin gene rearrangements (lymphoid malignancies).33 

Statistical analysis

Survival and progression-free survival were estimated by the Kaplan-Meier method, and mortality from various causes and GVHD were each summarized using cumulative incidence estimates.34The associations of chimerism values at day 28 with acute GVHD and rejection were examined using logistic regression. Associations of various patient characteristics with acute GVHD and rejection were examined using the Wilcoxon rank-sum test for continuous characteristics. The χ2 test was used for categorical characteristics, or Fisher's exact test if assumptions necessary for the χ2 test were not met. Data were analyzed as of April 1, 2000.

Table 2 summarizes the outcomes.

Table 2.

Outcomes after hematopoietic cell transplantation (grouped by rejection status and diagnoses)

UPNDiagnosis% donor CD3+RejectionDLIGVHDSurvival,
days
Summary of outcome as of 4/1/00
Day
28
Day
56
Acute (grade)Chronic
UL 6072-153 ALL-REL3 85 100  Yes*  3562-155 CR → REL → chemo/DLI → CR → Aspergillus 
 5772-153 MF-AML-IF 84 85   III Lim-S > 621 CR 
FH 142412-154 AML-aplasia 85 95   Ext-S, G > 549 CR 
 134612-153 AML-CR1 46 43  Yes × 2 II Ext-S > 759 Rel → chemo → CR → DLI → REL → chemo 
 145602-159 AML-CR1 40 99  Yes  > 473 CR 
UL 6462-153 AML-CR1 60 NE  Yes* II Lim-S > 341 Rel → DLI → CR → Rel → chemo/DLI → CR 
 6272-159 AML-CR1 NE NE    > 410 CR 
 6362-159 AML-CR1 58 63   III Lim-S > 383 CR 
UL 6162-159 AML-CR2 55 50  Yes*  1722-155 Rel → PD  
FH 133612-153 AML-CR3 50 90  Yes* II Ext-S, G 1682-155 Rel × 2 → DLI → CR mucor infection  
SU 18572-153 AML-IF 50 NE   NE 382-155 PD → conventional BMT  
FH 129142-153 CLL-REL2 100 100   II Ext-S, O 3612-155 MCR Pneumococcal sepsis 
SU 18792-153 CLL-PR2 56 86   III Ext-S, G > 528 MCR  
FH 145872-159 CLL-REF 10 20   Ext-S, L, O > 368 PR 
 145862-159 CLL-REF 90 80  Yes* II Ext-S, L 2442-155 PD  
SU 20282-159 CLL-PR2 30 25  Yes*  > 375 PR → PD  
FH 143222-159 CLL-REF 65 95   Ext-E, O, S > 319 MCR 
 146272-159 CLL-REF 71 96   III Ext-L, O, S > 325 CR  
UL 5962-153 CML-AP 79 95  Yes* II Lim-S > 530 MCR 
UL 6052-153 CML-CP 27 32   Lim-S > 488 MCR 
FH 142142-159 CML-CP 45 50   II Ext-O, S > 393 MCR  
UL 6342-159 CML-CP 43 70   Lim-O > 390 MCR 
FH 147262-159 CML-CP2-160 100 95   II Ext-E, G, L, O, S > 368 MCR  
SU 19552-153 HD-CR2 60 90  Yes × 2  1642-155 PD  
FH 138172-159 HD-PR 99 99   II Ext-E, G, O, S > 464 CR 
 142262-159 HD-REF 59 92  Yes* II Ext-S > 444 CR → Rel → XRT/chemo + DLI → PR 
 146922-159 HD-REF 70 90  Yes* II NE 1592-155 PD → Aspergillus infection 
 135272-159 MM-PR 99 97   II Ext-E, O, S > 416 CR 
 139222-159 MM-PR 83 94  Yes  > 417 CR2-164 
 119242-159 MM-PR 94 97    > 473 CR2-164 
 135412-159 MM-REF 98 100   II NE 912-155 PD 
 142742-159 MM-REF 90 95   II Ext-G, S > 310 CR  
UL 6252-159 MM-REF 100 NE   III NE 542-155 PR. Pneumonia, multiorgan failure  
FH 142242-159 NHL-REF 88 100   II NE 642-155 CR. Intracranial hemorrhage 
 144132-159 WM-REF 58 62   II Ext-E, O, S > 380 SD 
 146582-159 WM-REF 85 90  Yes × 2* Lim-S, O > 333 SD  
UL 6442-159 AML-CR1 20 Yes Yes  1332-155 Rel → 2nd NMHCT → engrafted → PD 
 142842-159 CLL-PR 19 24 Yes Yes  > 376 PD 
 140912-153 CML-AP 40 30 Yes Yes  2272-155 PD → Conventional BMT  
SU 20002-159 CML-AP 25 30 Yes Yes  4072-155 PD → 2nd NMHCT → rejected → PD  
SU 19482-153 CML-CP 60 30 Yes Yes  > 501 PD → 2nd NMHCT → rejected  
UL 6222-159 CML-CP 10 Yes Yes  > 341 PD 
FH 130092-153 MDS-RAEB-T 40 35 Yes Yes  1832-155 PD + liver cirrhosis 
 132462-153 MM-REF 43 35 Yes Yes  > 731 PD psoriasis in CR 
 132302-153 MM-PR 15 Yes Yes  > 751 REL 
UPNDiagnosis% donor CD3+RejectionDLIGVHDSurvival,
days
Summary of outcome as of 4/1/00
Day
28
Day
56
Acute (grade)Chronic
UL 6072-153 ALL-REL3 85 100  Yes*  3562-155 CR → REL → chemo/DLI → CR → Aspergillus 
 5772-153 MF-AML-IF 84 85   III Lim-S > 621 CR 
FH 142412-154 AML-aplasia 85 95   Ext-S, G > 549 CR 
 134612-153 AML-CR1 46 43  Yes × 2 II Ext-S > 759 Rel → chemo → CR → DLI → REL → chemo 
 145602-159 AML-CR1 40 99  Yes  > 473 CR 
UL 6462-153 AML-CR1 60 NE  Yes* II Lim-S > 341 Rel → DLI → CR → Rel → chemo/DLI → CR 
 6272-159 AML-CR1 NE NE    > 410 CR 
 6362-159 AML-CR1 58 63   III Lim-S > 383 CR 
UL 6162-159 AML-CR2 55 50  Yes*  1722-155 Rel → PD  
FH 133612-153 AML-CR3 50 90  Yes* II Ext-S, G 1682-155 Rel × 2 → DLI → CR mucor infection  
SU 18572-153 AML-IF 50 NE   NE 382-155 PD → conventional BMT  
FH 129142-153 CLL-REL2 100 100   II Ext-S, O 3612-155 MCR Pneumococcal sepsis 
SU 18792-153 CLL-PR2 56 86   III Ext-S, G > 528 MCR  
FH 145872-159 CLL-REF 10 20   Ext-S, L, O > 368 PR 
 145862-159 CLL-REF 90 80  Yes* II Ext-S, L 2442-155 PD  
SU 20282-159 CLL-PR2 30 25  Yes*  > 375 PR → PD  
FH 143222-159 CLL-REF 65 95   Ext-E, O, S > 319 MCR 
 146272-159 CLL-REF 71 96   III Ext-L, O, S > 325 CR  
UL 5962-153 CML-AP 79 95  Yes* II Lim-S > 530 MCR 
UL 6052-153 CML-CP 27 32   Lim-S > 488 MCR 
FH 142142-159 CML-CP 45 50   II Ext-O, S > 393 MCR  
UL 6342-159 CML-CP 43 70   Lim-O > 390 MCR 
FH 147262-159 CML-CP2-160 100 95   II Ext-E, G, L, O, S > 368 MCR  
SU 19552-153 HD-CR2 60 90  Yes × 2  1642-155 PD  
FH 138172-159 HD-PR 99 99   II Ext-E, G, O, S > 464 CR 
 142262-159 HD-REF 59 92  Yes* II Ext-S > 444 CR → Rel → XRT/chemo + DLI → PR 
 146922-159 HD-REF 70 90  Yes* II NE 1592-155 PD → Aspergillus infection 
 135272-159 MM-PR 99 97   II Ext-E, O, S > 416 CR 
 139222-159 MM-PR 83 94  Yes  > 417 CR2-164 
 119242-159 MM-PR 94 97    > 473 CR2-164 
 135412-159 MM-REF 98 100   II NE 912-155 PD 
 142742-159 MM-REF 90 95   II Ext-G, S > 310 CR  
UL 6252-159 MM-REF 100 NE   III NE 542-155 PR. Pneumonia, multiorgan failure  
FH 142242-159 NHL-REF 88 100   II NE 642-155 CR. Intracranial hemorrhage 
 144132-159 WM-REF 58 62   II Ext-E, O, S > 380 SD 
 146582-159 WM-REF 85 90  Yes × 2* Lim-S, O > 333 SD  
UL 6442-159 AML-CR1 20 Yes Yes  1332-155 Rel → 2nd NMHCT → engrafted → PD 
 142842-159 CLL-PR 19 24 Yes Yes  > 376 PD 
 140912-153 CML-AP 40 30 Yes Yes  2272-155 PD → Conventional BMT  
SU 20002-159 CML-AP 25 30 Yes Yes  4072-155 PD → 2nd NMHCT → rejected → PD  
SU 19482-153 CML-CP 60 30 Yes Yes  > 501 PD → 2nd NMHCT → rejected  
UL 6222-159 CML-CP 10 Yes Yes  > 341 PD 
FH 130092-153 MDS-RAEB-T 40 35 Yes Yes  1832-155 PD + liver cirrhosis 
 132462-153 MM-REF 43 35 Yes Yes  > 731 PD psoriasis in CR 
 132302-153 MM-PR 15 Yes Yes  > 751 REL 

GVHD indicates graft-versus-host disease; DLI, donor lymphocyte infusion; NE, not evaluable; PD, progressive disease; BMT, bone marrow transplantation; MCR, molecular remission; XRT, radiation therapy; SD, stable disease; NMHCT, nonmyeloablative hematopoietic cell transplantation; TBI, total-body irradiation; MMF, mycophenolate mofetil; and CSP, cyclosporine. For other abbreviations, see Table 1.

*

Indicates initial DLI given for relapse or progressive disease.

Indicates GVHD first occurred after DLI.

Indicates GVHD resolved; patient off immunosuppressive therapy. GVHD extent: Lim indicates limited GVHD; Ext, extensive GVHD. GVHD organ involvement: E indicates eyes; G, gastrointestinal; L, liver; O, oral; S, skin.

F2-153

Cohort 1: CSP scheduled day −1 to +35.

F2-155

Patient died.

F2-154

CSP taper delayed to day +100 to prevent GVHD because of active fungal infection.

F2-159

Cohort 2: CSP scheduled day −1 to +56.

F2-160

Patient 14726 received fludarabine 30 mg/m2 × 3 in addition to TBI/MMF/CSP.

F2-164

Trace monoclonal protein detected intermittently by immunofixation after CR was achieved.

Regimen-related toxicities and infections

No patient experienced regimen-related painful mucositis, severe nausea and vomiting, pulmonary toxicity, cardiac toxicity, hemorrhagic cystitis, or new-onset alopecia. Mild to moderate nausea and diarrhea due to MMF/CSP were common. Serum creatinine elevations to twice baseline values occurred in 59% of patients because of targeting high serum CSP levels, and resolved in most cases. One MM patient (UL625) with preexisting renal dysfunction (serum creatinine 2.3 mg/dL) required hemodialysis. Reversible hyperbilirubinemia to more than 10 mg/dL occurred in 3 patients because of preexisting liver cirrhosis (FH13009), concurrent amphotericin B treatment (FH14241), and isolated liver GVHD (FH14568). Documented bacterial infections requiring intravenous antibiotics occurred in 10 patients (22%) within 60 days after HCT. CMV reactivations were detected in 6 (24%) of 25 CMV-seropositive patients and were managed successfully with ganciclovir or foscarnet. Thirty-two patients were eligible for outpatient transplantation. Others had transplantation at the University of Leipzig (n = 12) or were already hospitalized (n = 1). Seventeen (53%) of the 32 patients did not require hospitalization within 60 days of HCT. The remainder were hospitalized for 1 to 35 days (median 8 days).

PB cell changes, allogeneic engraftment, and graft rejection

Figure 2 summarizes PB neutrophil and platelet changes to day +30. A median of 0 platelet units (range, 0-38 units) were transfused, and 34 patients (76%) required no transfusions. A median of 2 red blood cell units (range, 0-28 units) were transfused, and 14 patients (31%) required no transfusions. No serious hemolysis occurred in 14 ABO-incompatible transplants. Recipients of 6 major ABO-incompatible transplants (eg, A→O) required 0, 0, 0, 10, 23, and 28 red blood cell units.

Fig. 2.

Engraftment after nonmyeloablative HCT.

Engraftment profile showing neutrophil and platelet changes after HCT. Graphs show the median (black lines) and range (broken lines) of neutrophil and platelet counts of all 45 patients on days 0 through 30. ○, minimum and maximum values on each day.

Fig. 2.

Engraftment after nonmyeloablative HCT.

Engraftment profile showing neutrophil and platelet changes after HCT. Graphs show the median (black lines) and range (broken lines) of neutrophil and platelet counts of all 45 patients on days 0 through 30. ○, minimum and maximum values on each day.

Close modal

All patients had initial engraftment (Tables 2 and3). Based on T-cell engraftment, 86% were mixed chimeras and 14% were complete chimeras on day 28. This changed to 79% and 21%, respectively, on day 56. Nine (20%) of 44 patients given TBI/CSP/MMF had graft rejection between 2 and 4 months. Four had CML, 2 MM, one AML, one CLL, and one myelodysplasia. Rejections were nonfatal and usually associated with transient minor disturbances of blood counts (Figure 3), except for one patient (FH13246) who had 3 weeks of pancytopenia before recovery. This patient had experienced minimal myelosuppression until the onset of aplasia at 2 months after transplantation. Day-56 chimerism studies showed T cells to be 65% host and granulocytes to be 57% host at a time shortly before rejection when PB counts were not depressed, and these values were increased from 57% (T cells) and 29% (granulocytes) on day 28. Pretransplantation factors predictive of graft rejection were a lack of intensive preceding therapy and a diagnosis of CML (Table 4). Low donor T-cell chimerism at day 28 predicted rejections (Tables5,6). To reduce the risk of graft rejection, fludarabine 30 mg  ·  m−2  ·  d−1 on days −4, −3, and −2 was added to the regimen. In one patient (FH14726; CML) reported here, T cells at day 28 were 100% donor, as compared with 10% to 79% (median 42%) donor in 8 preceding CML patients.

Table 3.

Median percent and chimerism status

Day 283-150Day 563-151
Median % (range) of donor cells   
 T cells 60 (10-100) 91 (5-100)  
 PB neutrophils 91 (0-100) 99 (4-100)  
 Bone marrow 87 (0-100) 95 (2-100)  
Chimerism   
 % Mixed3-152 86 79  
 % Full donor3-153 14 21 
Day 283-150Day 563-151
Median % (range) of donor cells   
 T cells 60 (10-100) 91 (5-100)  
 PB neutrophils 91 (0-100) 99 (4-100)  
 Bone marrow 87 (0-100) 95 (2-100)  
Chimerism   
 % Mixed3-152 86 79  
 % Full donor3-153 14 21 
F3-150

There were 44 evaluable patients. One additional patient-donor pair (UL627) had no identifiable DNA polymorphism, and evidence of donor cell engraftment was based on red blood cell polymorphisms.

F3-151

There were 41 evaluable patients.

F3-152

At least 1 and no more than 95% T cells of donor origin.

F3-153

More than 95% T cells of donor origin.

Fig. 3.

Graft rejection after HCT.

Example of graft rejection after HCT, showing neutrophil and platelet changes in patient SU2000 with CML. Initial engraftment and subsequent rejection between 2 and 3 months were documented by chimerism studies (bottom panel). ▴, CD3+; ■, polymorphonuclear leukocytes (PMNs); ●, bone marrow (BM).

Fig. 3.

Graft rejection after HCT.

Example of graft rejection after HCT, showing neutrophil and platelet changes in patient SU2000 with CML. Initial engraftment and subsequent rejection between 2 and 3 months were documented by chimerism studies (bottom panel). ▴, CD3+; ■, polymorphonuclear leukocytes (PMNs); ●, bone marrow (BM).

Close modal
Table 4.

Rejection and graft-versus-host disease: analysis of pretransplantation factors

FactorRejectors (n = 9)Nonrejectors (n = 35)P value (rejection)Grades II-IV GVHD (n = 17)Grades 0-I GVHD (n = 19)P value (GVHD)
Male patient 7/9 (78%) 24/35 (67%) .70 11/17 (65%) 13/19 (68%) .81 
Male donor 4/9 (44%) 21/35 (58%) .47 9/17 (53%) 12/19 (63%) .54 
Intensive prior therapy4-150 1/9 (11%) 31/35 (86%) < .0001 15/17 (88%) 16/19 (84%) 1.0 
Donor age in years 48 (40-72) 57 (33-73) .80 53 (33-73) 58 (38-65) .78 
Patient age in years 54 (40-71) 56 (31-72) .76 56 (31-70) 57 (36-72) .87 
CD34+ cell dose × 106/kg 9.1 (3.8-15.1) 6.9 (3.1-17.5) .51 6.4 (3.3-16.1) 6.9 (3.1-17.5) .57 
CD3+ cell dose × 108/kg 3.3 (1.0-4.5) 3.4 (0.7-8.3) .58 3.6 (0.7-7.2) 3.1 (0.8-8.3) .86 
Diagnosis of CML 4/9 (44%) 4/35 (11%) .04 2/17 (12%) 3/19 (16%) 1.0 
FactorRejectors (n = 9)Nonrejectors (n = 35)P value (rejection)Grades II-IV GVHD (n = 17)Grades 0-I GVHD (n = 19)P value (GVHD)
Male patient 7/9 (78%) 24/35 (67%) .70 11/17 (65%) 13/19 (68%) .81 
Male donor 4/9 (44%) 21/35 (58%) .47 9/17 (53%) 12/19 (63%) .54 
Intensive prior therapy4-150 1/9 (11%) 31/35 (86%) < .0001 15/17 (88%) 16/19 (84%) 1.0 
Donor age in years 48 (40-72) 57 (33-73) .80 53 (33-73) 58 (38-65) .78 
Patient age in years 54 (40-71) 56 (31-72) .76 56 (31-70) 57 (36-72) .87 
CD34+ cell dose × 106/kg 9.1 (3.8-15.1) 6.9 (3.1-17.5) .51 6.4 (3.3-16.1) 6.9 (3.1-17.5) .57 
CD3+ cell dose × 108/kg 3.3 (1.0-4.5) 3.4 (0.7-8.3) .58 3.6 (0.7-7.2) 3.1 (0.8-8.3) .86 
Diagnosis of CML 4/9 (44%) 4/35 (11%) .04 2/17 (12%) 3/19 (16%) 1.0 

For abbreviations, see Tables 1 and 2.

Data are presented as n (%) or median (range). Rejection analysis includes 44 patients who received low-dose TBI/CSP/MMF and excludes patient FH14726, who also received fludarabine.

F4-150

Intensive prior therapy includes prior autograft, prior intensive chemotherapy for acute leukemia, or more than 3 cycles of a fludarabine-containing regimen.

Table 5.

Association of blood and marrow chimerism status at day 28 with subsequent rejection and acute graft-versus-host disease

Cell fractionPercent donor chimerism as a continuous (0-100%) variable
RejectionGVHD
OR5-15095% CIPvalue5-151OR5-15095% CIPvalue5-151
Marrow 0.4 0.2-0.8 .009 0.7 0.3-1.6 .42 
PB neutrophils 0.4 0.2-0.7 .004 1.2 0.6-2.5 .62 
PB T cells 0.1 0.04-0.5 .003 2.6 1.1-6.4 .03 
Cell fractionPercent donor chimerism as a continuous (0-100%) variable
RejectionGVHD
OR5-15095% CIPvalue5-151OR5-15095% CIPvalue5-151
Marrow 0.4 0.2-0.8 .009 0.7 0.3-1.6 .42 
PB neutrophils 0.4 0.2-0.7 .004 1.2 0.6-2.5 .62 
PB T cells 0.1 0.04-0.5 .003 2.6 1.1-6.4 .03 

Analysis of GVHD restricted to 35 patients with sustained engraftment and without GVHD before day 28. Patient UL627 was excluded from analysis because of lack of DNA polymorphism.

GVHD indicates graft-versus-host disease; CI, confidence interval; OR, odds ratio; PB, peripheral blood.

F5-150

OR presented as an increase of 25% in donor chimerism. For example, the odds of rejection among patients with 50% donor T-cell chimerism are 0.4 times the odds of rejection among patients with 25% donor T-cell chimerism.

F5-151

P value obtained from logistic regression, Wald test.

Table 6.

Association of donor T-cell chimerism with rejection and acute graft-versus-host disease

Percent donor PB T-cell chimerism as a categorical variable
% donor T cells on day 28No. rejection/no. at risk (%)No. GVHD/no. at risk (%)
0-25 5/6 (83%) 0/1 (0%) 
26-50 3/11 (27%) 2/8 (25%) 
51-75 1/11 (9%) 5/10 (50%) 
76-100 0/16 (0%) 9/15 (63%)  
Pvalue .00016-150 .326-150 
Percent donor PB T-cell chimerism as a categorical variable
% donor T cells on day 28No. rejection/no. at risk (%)No. GVHD/no. at risk (%)
0-25 5/6 (83%) 0/1 (0%) 
26-50 3/11 (27%) 2/8 (25%) 
51-75 1/11 (9%) 5/10 (50%) 
76-100 0/16 (0%) 9/15 (63%)  
Pvalue .00016-150 .326-150 

GVHD indicates graft-versus-host disease; PB, peripheral blood.

Analysis of GVHD restricted to 35 patients with sustained engraftment and without GVHD before day 28. Patient UL627 was excluded from analysis because of lack of DNA polymorphism.

F6-150

P value obtained from Fisher's exact test.

Patients with sustained allografts

Of 36 patients, 17 (47%) developed acute GVHD, which was grade II in 13 (36%) and grade III in 4 (11%) (Figure4A). The day-28 levels of donor T-cell chimerism predicted subsequent acute GVHD (Tables 5 and 6), which occurred in all patients with full donor chimerism. With one exception, acute GVHD began after discontinuation of MMF. In most cases, it responded to continuation of CSP and a standard course of methylprednisolone.35 Twenty-three (74%) of 31 evaluable patients developed chronic GVHD; 19 (61%) were treated with systemic immunosuppressive therapy, one with topical steroids, and 3 received no specific therapy.31 The extent of chronic GVHD and organ involvement are shown in Table 2. Of the patients treated for chronic GVHD, 9 had excellent responses, 9 had partial responses (PRs), and 2 were unevaluable because of death. Two patients discontinued therapy without a flare of GVHD.

Fig. 4.

GVHD and survival after HCT.

(A) Incidence of acute GVHD: cumulative incidence curves for patients with sustained engraftment. (B) Chronic GVHD: cumulative incidence curve for patients with sustained engraftment. Curve includes both chronic extensive and limited GVHD. (C) Survival, transplantation-related mortality, and relapse mortality after HCT. Survival estimates were 68.6% (95% CI = 54.9% to 82.2%) at 12 months and 65.0% (95% CI = 50.3% to 79.6%) at 18 months. Transplantation-related mortality (TRM) was regarded as a competing risk for death from relapse, relapse as a competing risk for TRM, and death without GVHD as a competing risk for GVHD. (D) Progression-free survival. Freedom from disease progression is shown among the entire cohort and among patients without graft rejection. Disease progression was defined as relapse from CR or disease progression that required additional chemotherapy and/or DLI after transplantation or > 25% increase in tumor measurements over those present at transplantation baseline.

Fig. 4.

GVHD and survival after HCT.

(A) Incidence of acute GVHD: cumulative incidence curves for patients with sustained engraftment. (B) Chronic GVHD: cumulative incidence curve for patients with sustained engraftment. Curve includes both chronic extensive and limited GVHD. (C) Survival, transplantation-related mortality, and relapse mortality after HCT. Survival estimates were 68.6% (95% CI = 54.9% to 82.2%) at 12 months and 65.0% (95% CI = 50.3% to 79.6%) at 18 months. Transplantation-related mortality (TRM) was regarded as a competing risk for death from relapse, relapse as a competing risk for TRM, and death without GVHD as a competing risk for GVHD. (D) Progression-free survival. Freedom from disease progression is shown among the entire cohort and among patients without graft rejection. Disease progression was defined as relapse from CR or disease progression that required additional chemotherapy and/or DLI after transplantation or > 25% increase in tumor measurements over those present at transplantation baseline.

Close modal

Status of underlying diseases: patients with graft rejection

Eight of 9 patients had persistent disease, and one MM patient (FH13230) achieved CR before relapse (Table 2). Patient FH13246 (MM) experienced a sustained CR of long-standing (30 years) psoriatic arthritis.

Status of underlying diseases: patients with sustained engraftment

Of 36 patients, 25 were alive, with 19 (53%) in CR, 2 in PR, and 4 without responses (Table 2). Twenty-nine of 36 patients had measurable disease before transplantation, and 19 (66%) achieved CR sometime after HCT. Progression-free survival for the entire group and survival for patients with sustained engraftment are shown in Figure 4C.

Five of 6 AML patients having transplantation in first CR or during aplasia were in remission. Of 2 AML patients in second or third remission, one died of relapse and one of infection while in remission. Of 3 patients with refractory acute leukemia, 2 died of relapse and one remained in remission at more than 23 months. All 5 CML patients were alive in molecular CR (Figure 5A). Of 7 CLL patients, 3 were in CR including 2 in molecular CR (Figure 5B), one was in PR, and one had progressive disease. One died of infection while in molecular CR and one died of progressive disease. Molecular CRs were first documented 5 to 12 months after transplantation and occurred gradually (Figure 5A,B). These remissions occurred in all 8 patients while they were receiving immunosuppressive therapy for management of GVHD. Of 6 MM patients, 4 were in CR (Figure 5C), one died of progressive disease, and one died of infection. Of 4 Hodgkin disease patients, one was in CR, one was alive in relapse, and 2 died of progressive disease. One NHL patient died of cerebral hemorrhage while in CR at 2 months. Two patients with Waldenström macroglobulinemia were alive with stable disease.

Fig. 5.

Complete disease responses after HCT.

(A) Example of molecular remission of CML (patient FH14726) induced by HCT without DLI, as documented by failure of reverse transcription–PCR to detect bcr-abl transcripts. The lane described “−ve” is a negative control of normal bone marrow (BM). K562 is a positive control for bcr-abl. (B) Example of molecular remission of CLL induced by HCT (patient FH12914) without DLI, as documented by PCR to detect a tumor-specific immunoglobulin heavy-chain gene rearrangement (arrow). Each posttransplantation sample was amplified in duplicate. The lanes designated 10−1 to 10−5 show a dilution series of the patient's pretransplantation sample (more than 90% tumor cells) into normal bone marrow. (C) Example of complete remission of MM (patient FH13922) after allogeneic HCT. The patient was initially treated with 4 cycles of vincristine, adriamycin, and dexamethasone (VAD). High-dose cytoreduction with melphalan 200 mg/m2 and autologous transplantation were performed 3 months before allogeneic HCT. DLI was given 4 months after HCT because of persistent tumor. After CR was achieved, trace levels of serum monoclonal paraprotein detected by immunofixation were present intermittently in follow-up testing.

Fig. 5.

Complete disease responses after HCT.

(A) Example of molecular remission of CML (patient FH14726) induced by HCT without DLI, as documented by failure of reverse transcription–PCR to detect bcr-abl transcripts. The lane described “−ve” is a negative control of normal bone marrow (BM). K562 is a positive control for bcr-abl. (B) Example of molecular remission of CLL induced by HCT (patient FH12914) without DLI, as documented by PCR to detect a tumor-specific immunoglobulin heavy-chain gene rearrangement (arrow). Each posttransplantation sample was amplified in duplicate. The lanes designated 10−1 to 10−5 show a dilution series of the patient's pretransplantation sample (more than 90% tumor cells) into normal bone marrow. (C) Example of complete remission of MM (patient FH13922) after allogeneic HCT. The patient was initially treated with 4 cycles of vincristine, adriamycin, and dexamethasone (VAD). High-dose cytoreduction with melphalan 200 mg/m2 and autologous transplantation were performed 3 months before allogeneic HCT. DLI was given 4 months after HCT because of persistent tumor. After CR was achieved, trace levels of serum monoclonal paraprotein detected by immunofixation were present intermittently in follow-up testing.

Close modal

Donor lymphocyte infusions

Twenty-three patients (51%) underwent the first DLI on days 56 to 326, including all 9 patients who subsequently rejected their grafts. Thus, DLIs given at 1 × 107 CD3+ cells/kg were unable to prevent progression to complete graft rejection. Twelve of 14 remaining patients had measurable disease. Four achieved CR, 5 had no response, and 3 were unevaluable because of concurrent other therapy (n = 2) or early death due to infection (FH14692). Three patients developed their first GVHD after DLI.

Survival and causes of death

With median follow-up at 417 days (range, 310-769 days), 30 of 45 patients (66.7%) were alive (Figure 4D). Twelve patients (26.7%) died of progressive disease, including 4 from DLI-induced GVHD and infections and 2 after conventional second HCT. Three (6.7%) died of transplantation complications without disease progression, including pneumonia at 2 months (UL625), central nervous system hemorrhage at 2 months (FH14224), and streptococcal sepsis after sinusitis at 1 year while off all systemic immunosuppression (FH12914).

This clinical protocol was a direct translation of preclinical canine HCT studies.22 The new concept for HCT, which relies on GVT effects rather than extreme chemoradiation to eradicate malignancies, has several advantages. First, serious organ damage characteristic of conventional transplantation is avoided. Second, a lack of profound pancytopenia reduces infection risks. These 2 features allow application of HCT to otherwise ineligible patients. Third, there may be an immunologic advantage that is the key to the success of this approach. Specifically, minimally cytotoxic pretransplantation conditioning allows persistence of host antigen-presenting cells, thus enhancing presentation of host minor antigens to donor T cells and initiation of GVT (and GVH) effects.

By exploiting postgrafting immunosuppression to control graft rejection, most of the pretransplantation TBI required for engraftment was eliminated in canine studies.22 Encouraged by the effectiveness and safety of these transplantations together with the therapeutic potential of donor lymphocytes,11-16 we initiated this study. Several conclusions are possible.

First, the regimen was safe and minimally toxic. Alopecia, severe mucositis, severe protracted pancytopenias, and fatal regimen-related toxicities typical of conventional HCT were not encountered. This allowed 53% of eligible patients to be treated entirely as outpatients, with others having relatively short hospitalizations (median 8 days), as compared with medians of more than 30 days with conventional HCT.36,37 Most patients required neither antibiotic treatment for infections nor platelet transfusions. Given the modest declines in blood counts, it was difficult to estimate the speed of engraftment. However, in one patient who was aplastic at transplantation (FH14241), neutrophils rose to more than 500/μL by day 8 and platelets to more than 20 000/μL by day 13.25Thus, the degree and exact tempo of myelosuppression induced by 200 cGy TBI in humans not given stem cell support cannot be estimated from this study. However, in dogs given this TBI dose without stem cell transplantation, the granulocyte nadir was 750/μL on day 20 and platelet nadirs were at 16 to 24 days after transplantation. Marrow recovery occurred in 17 of 18 dogs and all 17 survived beyond 30 days, with recovery complete by day 40 for granulocytes and by day 50 for platelets.22 CMV reactivations occurred in only 24% of CMV-seropositive patients, as compared with 79% after conventional transplantation.38 Three patients (6.7%) died of transplantation complications without disease progression. Overall, this toxicity profile was impressive considering the ages, preceding therapies, and preexisting organ dysfunction or infections among patients who were not conventional allografting candidates. These observations support the concept that the use of potent postgrafting immunosuppression permits one to reduce the intensity of pretransplantation immunosuppressive and myelosuppressive therapy, thereby reducing toxicities usually associated with allografting.

As with any radiation, there is a risk of late malignancies even from the low dose rate of 200 cGy TBI used here. Nevertheless, evidence suggests that this risk might be small. Among 95 673 nuclear workers exposed to low-level radiation, the excess relative risk of cancer was estimated to be only −0.02 (95% confidence interval [CI] −0.34 to 0.35) per sievert (approximately 1 Gy γ-radiation).39 In a large retrospective study, TBI-associated cancer risks after conventional HCT were dose related, but were not increased with TBI-based regimens using less than 10 Gy as compared with chemotherapy-only regimens.40 Long-term follow-up of individuals exposed to ionizing radiation after atomic-bomb explosions also supports the concept that the risks from low-dose TBI are relatively small.41 Others have reported a significant incidence of late leukemia or cancer after low-dose TBI when patients received fractionated high-dose-rate (150 cGy/min) TBI and, in most cases, additional high-dose local radiation and/or systemic alkylating agents.42 Based on those observations, it is difficult to estimate the late cancer risks from single-fraction low-dose-rate TBI as used here. The low dose rate used here (7 cGy/min), together with allografting, which largely eliminates host hematopoiesis through GVH reactions, should provide protection against secondary AML. Nonetheless, concerns about long-term safety have provided impetus to further reduce reliance on TBI as immunosuppression, particularly when treating nonmalignant disorders in younger patients. Additional canine studies have provided evidence that alternative, less toxic immunosuppression may eventually allow reduction or elimination of TBI from the current protocols.43 The risks of late malignancies from alternative reduced-intensity allografting regimens that use fludarabine with intermediate-dose or high-dose alkylating agents17-19 44 also remain uncertain and must be determined by longer follow-up.

Second, stable allogeneic engraftment was achieved in most patients despite using only 200 cGy TBI before transplantation. It is known from canine studies that 200 cGy is by itself not sufficiently immunosuppressive to permit stable allogeneic engraftment. However, when 200 cGy TBI before HCT was combined with postgrafting MMF/CSP, stable engraftment was seen in 11 of 12 dogs, whereas no long-term engraftment was seen with CSP alone. To achieve the same rate of engraftment without postgrafting immunosuppression, 920 cGy TBI was required. Put another way, MMF/CSP after grafting provided a degree of host immunosuppression that was otherwise achieved with more than 700 cGy TBI before transplantation. These considerations suggest that postgrafting CSP/MMF had an important role in facilitating engraftment in this clinical study. Nonfatal graft rejection occurred in 20% of the 44 patients who received TBI/MMF/CSP, and autologous hematopoietic recovery occurred in each case, as in the canine studies,22 providing evidence that host hematopoiesis was not ablated by the conditioning therapy. Rejection episodes occurred at 2 to 4 months, and one patient had 3 weeks of severe pancytopenia before recovery. The relatively large and increasing admixture of host cells, together with relatively normal PB counts shortly before rejection, suggests that TBI-induced suppression was not responsible for pancytopenia in this patient and implicates another mechanism, possibly similar to that observed with DLI-induced aplasia. The factor most closely associated with rejection was the absence of intensive previous chemotherapy. The current approach was most effective for heavily pretreated patients, with 30 of 31 (97%) having sustained engraftment. To reduce the rejection risk, we added the potent immunosuppressive purine analogue fludarabine to the pretransplantation conditioning for one patient reported here, and this approach is being evaluated in further studies. The high-level day-28 T-cell engraftment in CML patient FH14726 together with uniformly stable engraftment in 43 subsequent HLA-identical siblings (unpublished observations) and 11 unrelated donor HCT recipients45 suggests that fludarabine/TBI more effectively prevents rejection than TBI alone. Fludarabine synergizes with radiation to kill tumor cells,46 and this may also be true for its effect on T lymphocytes. It has been a component of most other reported reduced-intensity allografting conditioning regimens.17-19 44 

Third, 47% of patients with sustained engraftment developed acute grades II to III GVHD, with no grade IV disease. By comparison, rates of acute GVHD after conventional allografting using standard methotrexate/CSP prophylaxis47 were 35% and 44% among 44648 and 16449 younger patients, respectively, and 60% among 57 older patients (50-60 years) (R. Storb, unpublished data, Fred Hutchinson Cancer Research Center). The lack of stable mixed chimerism in most patients together with the development of GVHD in some patients after discontinuation of CSP on day 35 led us to extend CSP administration to day +56 for better GVHD protection. GVHD was effectively managed in most current patients. Whereas the median onset of acute GVHD was day 16 after conventional transplantation,48 the current median was 40 days. The almost complete absence of GVHD during days of combined MMF/CSP prophylaxis suggests that this is a potent immunosuppressive combination, consistent with canine GVHD studies.50 The contributions of low-intensity pretransplantation conditioning and initial mixed chimerism to the relatively benign courses of GVHD in these older patients remain unclear. Given the importance of inducing GVT effects for successful nonmyeloablative transplantation and the appreciable incidence of both acute and chronic GVHD encountered, GVHD control remains a critical research objective. Because MMF/CSP was given for relatively short periods after transplantation in this study, GVHD control might be improved simply by extending the duration of postgrafting immunosuppression.

Fourth, an important initial goal was establishing mixed chimerism, anticipating that it might protect from GVHD and it would provide an immunologic platform for subsequent DLI.51 Although approximately 80% of patients had mixed chimerism early after transplantation, it was usually unstable, progressing to either full donor chimerism or graft rejection and restricting DLI to 22 patients, including 8 who rejected their grafts. The differences between dogs and human patients with respect to establishing stable mixed chimerism may relate to differences in age and thymic function, previous chemotherapy and transfusions in patients, and minor interspecies variations in responses to the immunosuppressive agents.

Finally, persisting antitumor responses occurred exclusively in patients with sustained engraftment, indicating the importance of donor T cells for inducing and maintaining these responses. Nineteen of 29 patients (66%) with measurable disease before transplantation achieved CRs, and 3 others achieved PRs. Among these, 8 molecular remissions in CML (n = 5) and CLL (n = 3) patients were documented 5 to 12 months after HCT, and 7 of these occurred without the need for DLI. The molecular remissions occurred while patients received immunosuppression for treatment of GVHD, indicating that powerful GVT responses were generated and continued even with control of other GVHD manifestations. Although low-dose TBI given with or without alkylating agents may induce remissions in some patients with untreated CLL and NHL,52,53 it has not been reported that low-dose TBI can induce CRs and molecular remissions in patients with chemotherapy-resistant CLL, and in particular after failure of previous fludarabine-containing regimens. Similarly, although low-dose TBI may have transient suppressive effects on CML, even high-dose myeloablative regimens (at least 1200 cGy TBI plus intensive chemotherapy) are by themselves often incapable of providing sustained disease control, as indicated by the frequent relapses observed after using T-cell–depleted allografts.54 Thus, it is very unlikely that the low-dose and low-dose-rate TBI was responsible for the major disease responses observed here. Further, the molecular remissions occurred gradually, with clearing of tumor over many months and long after conditioning therapies were given. These observations also suggest that for some indolent malignancies, cytoreductive therapy given before transplantation may not be necessary and that DLI in this setting may be used sparingly and even avoided if disease is stable or responding. Factors that may influence the extent and tempo of disease responses include degrees of minor non-HLA disparities between donors and recipients, effects of immunosuppressive therapy given for GVHD, and expression of target antigens by tumor cells. For aggressive malignancies, it may be necessary to control tumor growth before GVT effects occur. Whereas others have used systemic chemotherapy, we are exploring whether this can be achieved by adding tumor-directed monoclonal antibody conjugates.55 56 

The induction of molecular remissions suggests that this treatment approach has curative potential for CML and CLL and may be a future alternative for younger patients who wish to avoid complications from high-dose regimens. Observations of durable molecular remissions of CML after DLI was given for relapse after conventional HCT support this notion.57 58 The current study included a heterogeneous group of patients, which allowed us to demonstrate antitumor effects in several different diseases and to identify patients at increased risk of graft rejection. Disease-specific phase 2 and 3 studies with prolonged follow-up will be required to fully evaluate the benefits and limitations of this approach regarding its curative potential, consequences of GVHD, late side effects, and comparisons with conventional allografting.

In smaller series reported recently, reduced-intensity regimens were used for allografting.17-20 Conceptually, achieving engraftment relied on immunosuppressive (and myelosuppressive) chemotherapy of varying intensities given before transplantation to suppress tumors and host immunity. Sustained engraftment was usually achieved, and impressive GVT activity was observed in some patients. Investigators from Houston used conventional chemotherapy regimens incorporating the purine analogues fludarabine or 2-chloro-deoxyadenosine in patients with advanced malignancies who were ineligible for conventional allografts.17,19 Postgrafting GVHD prophylaxis was omitted in their initial patients but added later to control GVHD. Fludarabine, 30 mg/m2 ×3, and cyclophosphamide, 300 mg/m2 ×3, their least intensive regimen, resulted in failed donor engraftment in 3 of 5 heavily pretreated CLL patients,19 necessitating dose escalation. These results suggest that this regimen was less immunosuppressive than TBI/MMF/CSP reported here. Others have used modifications of fludarabine/cyclophosphamide.44,59 Slavin et al18 reported using an attenuated conventional transplantation regimen of busulfan, fludarabine, and antithymocyte globulin with CSP alone as GVHD prophylaxis. Among 26 patients, veno-occlusive disease occurred in 50% and fatal acute GVHD occurred in 15%. Although the 14-month survival of 77.5% was impressive, the median patient age was 32 years (range, 1-56 years), and all appeared to be candidates for conventional allografting. Using high-dose cyclophosphamide and antithymocyte globulin with or without thymic irradiation, Sykes et al20 have established grafts from HLA-matched and HLA-mismatched donors in patients with advanced lymphoma.

By focusing on postgrafting immunosuppression to control rejection and GVHD, we have further reduced the intensity and toxicity of conditioning therapy for recipients of HLA-identical grafts. This allografting approach appears broadly applicable and suitable for studies of adoptive immunotherapy in hematologic and solid tumors, given that the transplantations can be performed effectively in an outpatient setting with minimal toxicity and limited supportive-care requirements. The development of effective targeting strategies to focus T-cell reactions against tumor antigens or minor antigens with hematopoietic restriction60,61 might enhance future outcomes through more effective tumor killing and by reducing the risks of GVHD. As reviewed elsewhere,51 this new allografting concept also has potentially important applications in the treatment of some nonmalignant diseases.

We thank the medical, nursing, data processing, and clinical staffs at the listed institutions for their important contributions to this study through their careful and dedicated care of the patients. We thank Bonnie Larson for her assistance in preparation of this manuscript.

Abbreviations for therapies listed in Table 1: ABVD indicates adriamycin, bleomycin, vinblastine, dacarbazine; Ara-C, cytosine arabinoside; Asp, asparaginase; CE ± D, high-dose cyclophosphamide, etoposide ± decadron; Ch, chlorambucil; CHAD, cisplatin, cytosine arabinoside, dexamethasone; CHOP, vincristine, adriamycin, cyclophosphamide, prednisone; CMOPP, cyclophosphamide, vincristine, procarbazine, prednisone; Cp, cisplatin; CVP, cyclophosphamide, vincristine, prednisone; Cy, cyclophosphamide; CyTx, high-dose cyclophosphamide, Taxol; D, daunorubicin; Dex, dexamethasone; DHAP, dexamethasone, cytosine arabinoside, cisplatin; E, etoposide; F, fludarabine; FLAG, fludarabine, cytosine arabinoside, granulocyte colony-stimulating factor; FMD, fludarabine, mitoxantrone, dexamethasone; Gc, gemcitabine; HAM, high-dose cytosine arabinoside, mitoxantrone; HDAC, high-dose cytosine arabinoside; HDCy, high-dose cyclophosphamide; HDT, high-dose chemotherapy and autologous transplantation; Hh, homoharringtonine; Hu, hydroxyurea; Ida, idarubicin; IFN, alpha-interferon; M, melphalan; MCE, mitoxantone, cytosine arabinoside, etoposide; Mi, mitoxantrone; MOPP, nitrogen mustard, vincristine, procarbazine, prednisone; P, prednisone; TAD, thioguanine, cytosine arabinoside, daunorubicin; TG, 6-thioguanine; V, vincristine; VAD, vincristine, adriamycin, dexamethasone; Vb, vinblastine; XRT, local radiation therapy.

Supported in part by grants CA78902, CA49605, HL36444, CA18221, CA15704, and HL03701 awarded by the National Institutes of Health, Department of Health and Human Services, Bethesda, MD. P.A.M. and D.G.M. were also supported by a grant from the Gabrielle Rich Leukemia Foundation. R.F.S. also received support from the Laura Landro Salomon Endowment Fund.

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
Molina
AJ
Storb
RF
Hematopoietic stem cell transplantation in older adults.
Handbook of Bone Marrow Transplantation. London
Rowe
JM
Lazarus
HM
Carella
AM
2000
111
137
Martin Dunitz Ltd
United Kingdom
2
Burchenal
JH
Oettgen
HF
Holmberg
EAD
Hemphill
SC
Reppert
JA
Effect of total body irradiation on the transplantability of mouse leukemias.
Cancer Res.
20
1960
425
430
3
Thomas
ED
Storb
R
Clift
RA
et al
Bone-marrow transplantation.
N Engl J Med.
292
1975
832
843
895-902.
4
Weiden
PL
Flournoy
N
Thomas
ED
et al
Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts.
N Engl J Med.
300
1979
1068
1073
5
Weiden
PL
Sullivan
KM
Flournoy
N
Storb
R
Thomas
ED
the Seattle Marrow Transplant Team
Antileukemic effect of chronic graft-versus-host disease. Contribution to improved survival after allogeneic marrow transplantation.
N Engl J Med.
304
1981
1529
1533
6
Horowitz
MM
Gale
RP
Sondel
PM
et al
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood.
75
1990
555
562
7
Butturini
A
Bortin
MM
Gale
RP
Graft-versus-leukemia following bone marrow transplantation.
Bone Marrow Transplant.
2
1987
233
242
8
Kersey
JH
Weisdorf
D
Nesbit
ME
et al
Comparison of autologous and allogeneic bone marrow transplantation for treatment of high-risk refractory acute lymphoblastic leukemia.
N Engl J Med.
317
1987
461
467
9
Weisdorf
DJ
Nesbit
ME
Ramsay
NKC
et al
Allogeneic bone marrow transplantation for acute lymphoblastic leukemia in remission: prolonged survival associated with acute graft-versus-host disease.
J Clin Oncol.
5
1987
1348
1355
10
Sullivan
KM
Weiden
PL
Storb
R
et al
Influence of acute and chronic graft-versus-host disease on relapse and survival after bone marrow transplantation from HLA-identical siblings as treatment of acute and chronic leukemia.
Blood.
73
1989
1720
1728
11
Kolb
HJ
Mittermüller
J
Clemm
CH
et al
Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients.
Blood.
76
1990
2462
2465
12
Porter
DL
Roth
MS
McGarigle
C
Ferrara
JLM
Antin
JH
Induction of graft-versus-host disease as immunotherapy for relapsed chronic myeloid leukemia.
N Engl J Med.
330
1994
100
106
13
Kolb
HJ
Schattenberg
A
Goldman
JM
et al
Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia.
Blood.
86
1995
2041
2050
14
Mackinnon
S
Papadopoulos
EB
Carabasi
MH
et al
Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease.
Blood.
86
1995
1261
1268
15
Giralt
S
Hester
J
Huh
Y
et al
CD8-depleted donor lymphocyte infusion as treatment for relapsed chronic myelogenous leukemia after allogeneic bone marrow transplantation.
Blood.
86
1995
4337
4343
16
Collins
RH
Jr
Shpilberg
O
Drobyski
WR
et al
Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation.
J Clin Oncol.
15
1997
433
444
17
Giralt
S
Estey
E
Albitar
M
et al
Engraftment of allogeneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft-versus-leukemia without myeloablative therapy.
Blood.
89
1997
4531
4536
18
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
19
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
20
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
21
Storb
R
Raff
RF
Appelbaum
FR
et al
Comparison of fractionated to single-dose total body irradiation in conditioning canine littermates for DLA-identical marrow grafts.
Blood.
74
1989
1139
1143
22
Storb
R
Yu
C
Wagner
JL
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
23
Storb
R
Raff
RF
Graham
T
et al
Marrow toxicity of fractionated versus single dose total body irradiation is identical in a canine model.
Int J Radiat Oncol Biol Phys.
26
1993
275
283
24
Mickelson
E
Petersdorf
EW
Histocompatibility.
Hematopoietic Cell Transplantation.
2nd ed.
Thomas
ED
Blume
KG
Forman
SJ
1999
28
37
Blackwell Science
Boston
25
Xun
CQ
McSweeney
PA
Boeckh
M
Storb
RF
Broudy
VC
Thompson
JA
Successful nonmyeloablative allogeneic hematopoietic stem cell transplant in an acute leukemia patient with chemotherapy-induced marrow aplasia and progressive pulmonary aspergillosis [letter].
Blood.
94
1999
3273
3276
26
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
27
Slavin
MA
Osborne
B
Adams
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
28
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
29
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
30
Przepiorka
D
Weisdorf
D
Martin
P
et al
Consensus conference on acute GVHD grading.
Bone Marrow Transplant.
15
1995
825
828
31
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
32
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
33
Radich
JP
Gehly
G
Gooley
T
et al
Polymerase chain reaction detection of the BCR-ABL fusion transcript after allogeneic marrow transplantation for chronic myeloid leukemia: results and implications in 346 patients.
Blood.
85
1995
2632
2638
34
Gooley
TA
Leisenring
W
Crowley
J
Storer
BE
Estimation of failure probabilities in the presence of competing risks: new representations of old estimators.
Stat Med.
18
1999
695
706
35
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
36
Storb
R
Pepe
M
Anasetti
C
et al
What role for prednisone in prevention of acute graft-versus-host disease in patients undergoing marrow transplants?
Blood.
76
1990
1037
1045
37
Storb
R
Etzioni
R
Anasetti
C
et al
Cyclophosphamide combined with antithymocyte globulin in preparation for allogeneic marrow transplants in patients with aplastic anemia.
Blood.
84
1994
941
949
38
Boeckh
M
Gooley
TA
Myerson
D
Cunningham
T
Schoch
G
Bowden
RA
Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir versus ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study.
Blood.
88
1996
4063
4071
39
Cardis
E
Gilbert
ES
Carpenter
L
et al
Effects of low doses and low dose rates of external ionizing radiation: cancer mortality among nuclear industry workers in three countries.
Radiat Res.
142
1995
117
132
40
Curtis
RE
Rowlings
PA
Deeg
HJ
et al
Solid cancers after bone marrow transplantation.
N Engl J Med.
336
1997
897
904
41
Thompson
DE
Mabuchi
K
Ron
E
et al
Cancer incidence in atomic bomb survivors. Part II: Solid tumors, 1958-1987 [review].
Radiat Res.
137(suppl 2)
1994
S17
S67
42
Travis
LB
Weeks
J
Curtis
RE
et al
Leukemia following low-dose total body irradiation and chemotherapy for non-Hodgkin's lymphoma.
J Clin Oncol.
14
1996
565
571
43
Storb
R
Yu
C
Zaucha
JM
et al
Stable mixed hematopoietic chimerism in dogs given donor antigen, CTLA4Ig, and 100 cGy total body irradiation before and pharmacologic immunosuppression after marrow transplant.
Blood.
94
1999
2523
2529
44
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
45
Niederwieser
D
Wolff
D
Hegenbart
U
et al
Hematopoietic stem cell transplants (HSCT) from HLA-matched and one-allele mismatched unrelated donors using a nonmyeloablative regimen [abstract #2506].
Blood.
94(suppl 1)
1999
561a
46
Gregoire
V
Hunter
N
Milas
L
Brock
WA
Plunkett
W
Hittelman
WN
Potentiation of radiation-induced regrowth delay in murine tumors by fludarabine.
Cancer Res.
54
1994
468
474
47
Deeg
HJ
Shulman
HM
Schmidt
E
Yee
GC
Thomas
ED
Storb
R
Marrow graft rejection and veno-occlusive disease of the liver in patients with aplastic anemia conditioned with cyclophosphamide and cyclosporine.
Transplantation.
42
1986
497
501
48
Nash
RA
Pepe
MS
Storb
R
et al
Acute graft-versus-host disease: analysis of risk factors after allogeneic marrow transplantation and prophylaxis with cyclosporine and methotrexate.
Blood.
80
1992
1838
1845
49
Ratanatharathorn
V
Nash
RA
Przepiorka
D
et al
Phase III study comparing methotrexate and tacrolimus (Prograf, FK506) with methotrexate and cyclosporine for graft-versus-host-disease prophylaxis after HLA-identical sibling bone marrow transplantation.
Blood.
92
1998
2303
2314
50
Yu
C
Seidel
K
Nash
RA
et al
Synergism between mycophenolate mofetil and cyclosporine in preventing graft-versus-host disease among lethally irradiated dogs given DLA-nonidentical unrelated marrow grafts.
Blood.
91
1998
2581
2587
51
McSweeney
PA
Storb
R
Mixed chimerism: preclinical studies and clinical applications [review].
Biol Blood Marrow Transplant.
5
1999
192
203
52
Roncadin
M
Arcicasa
M
Zagonel
V
et al
Total body irradiation and prednimustine in chronic lymphocytic leukemia and low grade non-Hodgkin's lymphomas: a 9-year experience at a single institution.
Cancer.
74
1994
978
984
53
Mendenhall
NP
Noyes
WD
Million
RR
Total body irradiation for stage II-IV non-Hodgkin's lymphoma: ten-year follow-up.
J Clin Oncol.
7
1989
67
74
54
Marmont
AM
Horowitz
MM
Gale
RP
et al
T-cell depletion of HLA-identical transplants in leukemia.
Blood.
78
1991
2120
2130
55
Sievers
EL
Appelbaum
FR
Spielberger
RT
et al
Selective ablation of acute myeloid leukemia using antibody-targeted chemotherapy: a phase I study of an anti-CD33 calicheamicin immunoconjugate.
Blood.
93
1999
3678
3684
56
Matthews
DC
Appelbaum
FR
Eary
JF
et al
Phase I study of 131I-Anti-CD45 antibody plus cyclophosphamide and total body irradiation for advanced acute leukemia and myelodysplastic syndrome.
Blood.
94
1999
1237
1247
57
Porter
DL
Collins
RH
Jr
Shpilberg
O
et al
Long-term follow-up of patients who achieved complete remission after donor leukocyte infusions.
Biol Blood Marrow Transplant.
5
1999
253
261
58
Cwynarski
K
Dazzi
F
Cross
NCP
et al
Response to donor lymphocyte infusions (DLI) of patients with CML in relapse after allografting: a long-term follow-up study using RT-PCR [abstract #2968].
Blood.
94(suppl 1)
1999
669a
59
Carella
AM
Lerma
E
Dejana
A
et al
Engraftment of HLA-matched sibling hematopoietic stem cells after immunosuppressive conditioning regimen in patients with hematologic neoplasias.
Haematologica.
83
1998
904
909
60
Warren
EH
Greenberg
PD
Riddell
SR
Cytotoxic T-lymphocyte-defined human minor histocompatibility antigens with a restricted tissue distribution.
Blood.
91
1998
2197
2207
61
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

Peter A. McSweeney, Bone Marrow Transplant Program, University of Colorado Health Sciences Center, 4200 East 9th Avenue, Campus Box B-190, Denver, CO 80262; e-mail:peter.mcsweeney@uchsc.edu.

Sign in via your Institution