Abstract
Purpose
We designed a minimal-intensity conditioning regimen for allogeneic hematopoietic cell transplantation (HCT) in patients with advanced hematologic malignancies unable to tolerate high-intensity regimens because of age, serious comorbidities, or previous high-dose HCT. The regimen allows the purest assessment of graft-versus-tumor (GVT) effects apart from conditioning and graft-versus-host disease (GVHD) not augmented by regimen-related toxicities.
Patients and Methods
Patients received low-dose total-body irradiation ± fludarabine before HCT from HLA-matched related (n = 611) or unrelated (n = 481) donors, followed by mycophenolate mofetil and a calcineurin inhibitor to aid engraftment and control GVHD. Median patient age was 56 years (range, 7 to 75 years). Forty-five percent of patients had comorbidity scores of ≥ 3. Median follow-up time was 5 years (range, 0.6 to 12.7 years).
Results
Depending on disease risk, comorbidities, and GVHD, lasting remissions were seen in 45% to 75% of patients, and 5-year survival ranged from 25% to 60%. At 5 years, the nonrelapse mortality (NRM) rate was 24%, and the relapse mortality rate was 34.5%. Most NRM was a result of GVHD. The most significant factors associated with GVHD-associated NRM were serious comorbidities and grafts from unrelated donors. Most relapses occurred early while the immune system was compromised. GVT effects were comparable after unrelated and related grafts. Chronic GVHD, but not acute GVHD, further increased GVT effects. The potential benefit associated with chronic GVHD was outweighed by increased NRM.
Conclusion
Allogeneic HCT relying on GVT effects is feasible and results in cures of an appreciable number of malignancies. Improved results could come from methods that control progression of malignancy early after HCT and effectively prevent GVHD.
INTRODUCTION
Allogeneic graft-versus-leukemia effects were first suggested by Barnes et al1 in 1956 after studying a murine model of syngeneic and H-2–incompatible hematopoietic cell transplantation (HCT). Reports linking graft-versus-host disease (GVHD) to antileukemic effects in humans were published many years later.2–5 Importantly, the International Bone Marrow Transplant Registry study4 showed that allogeneic recipients with little or no GVHD experienced less relapse than identical twin recipients, suggesting antileukemic effects even in the absence of clinical GVHD. Subsequently, donor lymphocyte infusions have been used to control leukemic relapse after HCT,6 and graft-versus-tumor (GVT) effects have been described for other hematologic malignancies (reviewed in Roddie and Peggs7).
We developed a minimal-intensity conditioning regimen for allogeneic HCT that includes low-dose total-body irradiation (TBI) with or without fludarabine.8 Postgrafting immunosuppression with mycophenolate mofetil (MMF) and a calcineurin inhibitor serves to aid engraftment and control GVHD. The approach was designed for patients with advanced hematologic malignancies who could not tolerate high-dose conditioning regimens historically used for HCT because of age, comorbidities, or unsuccessful prior HCT with high-dose conditioning. The use of a minimal-intensity conditioning regimen allows for the purest assessment of GVT effects apart from conditioning and the best determination of GVHD not augmented by regimen-related toxicities. The current retrospective analysis of data in 1,092 patients has enabled a comprehensive assessment of the success and limitations of this approach.
PATIENTS AND METHODS
Patients
Between December 16, 1997, and October 30, 2009, 1,092 consecutive patients with hematologic malignancies were entered onto prospective, multicenter trials registered with ClinicalTrials.gov (Appendix Table A1). The institutional review board at each collaborating center approved the trials. Patients signed institutional review board–approved consent forms.
Table 1 lists patient characteristics. Grafts consisted of granulocyte colony-stimulating factor-mobilized blood mononuclear cells containing medians of 7.8 × 106 CD34+ cells/kg (range, 0.8 to 42.6 × 106 CD34+ cells/kg) and 3.1 × 108 CD3+ cells/kg (range, 0.2 to 68.0 × 108 CD3+ cells/kg). Patients had received a median of three prior chemotherapy regimens (range, zero to 18 regimens). HCT comorbidity index (HCT-CI) scores were assigned as described.9 Forty-five percent of patients had serious comorbidities with scores of 3 and higher.
Table 1.
Patient Demographics and Clinical Characteristics
| Characteristic | Patients |
|||||
|---|---|---|---|---|---|---|
| All (N = 1,092) |
Related Recipientsa(n = 611) |
Unrelated Recipientsb(n = 481) |
||||
| No. | % | No. | % | No. | % | |
| Sex | ||||||
| Female | 402 | 37 | 226 | 37 | 176 | 37 |
| Male | 690 | 63 | 385 | 63 | 305 | 63 |
| Age, years | ||||||
| 0-19 | 15 | 1 | 3 | < 1 | 12 | 2 |
| 20-29 | 34 | 3 | 16 | 3 | 18 | 4 |
| 30-39 | 67 | 6 | 41 | 7 | 26 | 5 |
| 40-49 | 167 | 15 | 117 | 19 | 50 | 10 |
| 50-59 | 424 | 39 | 241 | 39 | 183 | 38 |
| 60-69 | 348 | 32 | 170 | 28 | 178 | 37 |
| 70-79 | 37 | 3 | 23 | 4 | 14 | 3 |
| Median | 56 | 56 | 58 | |||
| Range | 7-75 | 17-74 | 7-75 | |||
| Diagnosis | ||||||
| ALL | 46 | 4 | 11 | 2 | 35 | 7 |
| AML | 289 | 26 | 127 | 21 | 162 | 34 |
| CLL | 122 | 11 | 67 | 11 | 55 | 11 |
| CML | 47 | 4 | 26 | 4 | 21 | 4 |
| HL | 61 | 6 | 41 | 7 | 20 | 4 |
| MDS | 92 | 8 | 47 | 8 | 45 | 9 |
| MPN | 18 | 2 | 7 | 1 | 11 | 2 |
| MM | 217 | 20 | 170 | 28 | 47 | 10 |
| NHL | 190 | 17 | 109 | 18 | 81 | 17 |
| Waldenström's syndrome | 10 | 1 | 6 | 1 | 4 | 1 |
| Regimen | ||||||
| TBI 2 Gy | 222 | 20 | 222 | 36 | — | — |
| TBI 2 Gy + fludarabinec | 840 | 77 | 378 | 62 | 462 | 96 |
| TBId 3 Gy + fludarabine | 26 | 2 | 10 | 2 | 16 | 3 |
| TBId 4 Gy + fludarabine | 4 | < 1 | 1 | < 1 | 3 | 1 |
| Prior transplantation | ||||||
| None | 657 | 60 | 345 | 56 | 316 | 66 |
| Planned autologouse | 214 | 20 | 164 | 27 | 50 | 10 |
| Failed autologous | 174 | 16 | 87 | 14 | 87 | 18 |
| Other autologousf | 22 | 2 | 8 | 1 | 10 | 2 |
| Failed allogeneic | 25 | 2 | 7 | 1 | 18 | 4 |
| Year of transplantation | ||||||
| 1997-1999 | 71 | 6 | 71 | 12 | — | — |
| 2000-2004 | 601 | 55 | 344 | 56 | 257 | 53 |
| 2005-2009 | 423 | 39 | 196 | 32 | 224 | 47 |
| Follow-up of surviving patients, yearsg | ||||||
| Median | 5 | 5.1 | 5 | |||
| Range | 0.6 to 12.7 | 0.6 to 12.7 | 0.6 to 10.8 | |||
| HCT-comorbidity index scoresh | ||||||
| 0 | 210 | 24 | 124 | 27 | 86 | 20 |
| 1 | 107 | 12 | 54 | 12 | 53 | 12 |
| 2 | 168 | 19 | 85 | 19 | 83 | 19 |
| 3 | 209 | 24 | 104 | 23 | 105 | 24 |
| 4 | 80 | 9 | 34 | 7 | 46 | 11 |
| 5 | 55 | 6 | 25 | 6 | 30 | 7 |
| 6 | 38 | 4 | 18 | 4 | 20 | 5 |
| 7+ | 22 | 2 | 10 | 2 | 12 | 3 |
| CMV positivei | ||||||
| Patient | 641 | 60 | 374 | 63 | 267 | 56 |
| Donor | 485 | 46 | 339 | 58 | 146 | 31 |
NOTE. Fifty-five percent of patients were enrolled at Fred Hutchinson Cancer Research Center and 45% at collaborating centers. Donors and recipients were matched for HLA-A, -B, and -C, DRB1, and DQB1 by high-resolution typing except for 54 unrelated donor-recipient pairs.
Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; CML, chronic myelocytic leukemia; CMV, cytomegalovirus; HCT, hematopoietic cell transplantation; HL, Hodgkin lymphoma; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasms; MM, multiple myeloma; NHL, non-Hodgkin lymphoma; TBI, total-body irradiation.
Five phenotypic HLA matches.
Fifty-four mismatches for one HLA class I allele. In addition to mycophenolate mofetil/tacrolimus, 61 unrelated recipients received sirolimus as part of a randomized trial.
Fludarabine was added to TBI to reduce the risk of graft rejection.
Thirty patients with advanced myeloid malignancies received 3 Gy (n = 26) or 4 Gy (n = 4) TBI; even at these doses, TBI is nonmyeloablative, sublethal, and well tolerated.
Including one syngeneic transplantation.
Transplantation for other malignancy and now referred for secondary MDS/AML.
Data reflect events as of the date of last contact before the database was locked on March 1, 2011.
Missing for 203 patients.
Missing for 24 patients and 45 donors.
Conditioning and Postgrafting Immunosuppression
Patients received low-dose TBI with (n = 840) or without (n = 222) fludarabine 30 mg/m2 per day on days 4, 3, and 2 before HCT (Table 1). Immunosuppression after HCT included MMF (28 days for related8 and at least 96 days for unrelated10 recipients) and a calcineurin inhibitor (180 days), either cyclosporine (CSP; n = 814) or tacrolimus (TAC; n = 278). In our experience, CSP and TAC were equally effective for GVHD prevention.11 Infection prophylaxis and treatment followed institutional standard practice guidelines.
Monitoring After Transplantation
Patients had marrow aspirations on days 28, 84, 180, and 360, and then annually to assess disease status. Donor chimerism was evaluated on days 28, 84, and 365 after HCT. Acute and chronic GVHD were graded as described.12,13 Relapse was defined as recurrence of malignancy based on one or more of the following: marrow morphology, flow cytometry, cytogenetics including fluorescence in situ hybridization, electrophoresis, immunofixation assays, polymerase chain reaction–based assays for disease markers, or imaging results. Disease progression was based on at least 50% increase in disease burden.14 Donor lymphocyte infusions were given to 33 patients for relapse or progression and to 19 patients for declining donor chimerism.
Causes of Death
Relapse mortality included deaths after relapse or progression of disease present before transplantation, regardless of other events. Nonrelapse mortality (NRM) included all deaths without relapse or progression. GVHD mortality included all deaths in patients with a history of GVHD who died from infections or organ failure during immunosuppressive therapy. Infection was listed as cause of death in patients without relapse or progression or a history of GVHD.
Statistical Analysis
Survival was estimated using the Kaplan-Meier method.15 Rates of acute and chronic GVHD, relapse or progression, and NRM were estimated according to standard methods.16 Death was treated as a competing risk for all other end points. Relapse was treated as a competing risk for NRM. Cox regression analysis was used for risk factor analysis and for comparisons between groups defined by donor relation and other characteristics. Risk factors for NRM occurring with a history of grade 2 to 4 acute GVHD or chronic GVHD were evaluated using a stepwise backward elimination procedure. All candidate risk factors were included in the initial model, and the least significant factors were excluded in sequential fashion until all remaining factors were significant at the P = .05 level. Candidate risk factors included HCT-CI scores of ≥ 3, unrelated transplantation, recipient cytomegalovirus (CMV) seropositivity, prior cytoreductive autologous HCT, transplanted CD34 and CD3 cell doses, allele-level HLA class I mismatch, grafts from female donors into male recipients, the addition of fludarabine to TBI, CSP versus TAC, and recipient age. Missing HCT-CI scores and CMV serostatus were accommodated by including missing categories for these variables. Relapse rates per patient year were calculated by dividing the total number of relapse events by the total years of follow-up in the time period of interest. Follow-up in a time period was truncated by the earliest occurrence of relapse, death, last contact, or the end of the time period.
RESULTS
Engraftment/Chimerism
Sustained engraftment was documented in 96% of patients, whereas 4% experienced graft rejection or failure between days 13 and 1,123 (median, 82 days). Engraftment was prompt, as evidenced by increasing neutrophil and platelet counts following the nadirs after conditioning. On days 28, 84, and 365, the median proportions of donor-derived cells were 95%, 99%, and 100% in the marrow; 98%, 100%, and 100% in neutrophils in blood; and 84%, 90%, and 100% in CD3+ T cells in blood, respectively. Forty-four percent of patients were never hospitalized at any time, whereas 56% had a median of 10 hospitalization days (range, 1 to 233 days), including the single overnight hospitalization for infusion of unrelated grafts.
Among the 48 patients with graft rejection (n = 41) or graft failure (n = 7), 36 had myeloid malignancies, and 13 received TBI (2 Gy) alone. Of the 48 patients, 36 died between 49 and 3,936 days (median, 284 days) after HCT, 29 of relapse and seven of nonrelapse causes. Twelve patients are alive and in remission, seven after second HCT, two after treatment with pentostatin immunosuppression and donor lymphocyte infusion, and three, all with chronic myelocytic leukemia, after successful treatment with tyrosine kinase inhibitors.
Relapse or Progression
By 5 years, 43.5% of the 1,092 patients had disease relapse or progression (related recipients, 45%; unrelated recipients, 42%), and 34.5% had died from relapse. Among 98 patients alive at 5 years after relapse or progression, 58% had stable complete remission after discontinuing immunosuppression, after chemotherapy with or without donor lymphocyte infusion, or after another allogeneic HCT; 5% were in very good partial remission; 5% were in molecular relapse (chronic myelocytic leukemia); 12% had stable disease; 5% had progression; and the remaining 15% were unknown. Relapse rates varied depending on underlying diseases and disease stages (Table 2). The designation of low, standard, or high relapse risk for each disease and disease stage followed a previously described algorithm.14 The lowest risk group included patients with B-cell malignancies, especially non-Hodgkin lymphoma (NHL) at any stage except for aggressive NHL not in remission. The highest risk group included patients with advanced malignancies not in remission. All 18 patients with acute lymphoblastic leukemia (ALL) in ≥ second complete remission experienced relapse. Relapse rates were highest during the first 2 years after HCT, whereas rates during years 3 to 5 were low, except among patients with multiple myeloma (MM) or Hodgkin lymphoma (HL) not in remission at the time of HCT. The cumulative incidence rates of relapse or progression in the three risk groups followed the expected pattern (Fig 1). Median days until relapse in the low-, standard-, and high-risk groups were 182, 174, and 107, respectively.
Table 2.
Relapse Rates Per Patient Year Among 1,092 Patients
| Diagnosis* | Stage | No. of Patients | Relapse Rate |
|
|---|---|---|---|---|
| Years 1 and 2 | Years 3-5 | |||
| Low risk | ||||
| MPN | Any | 18 | 0.10 | 0.00 |
| CLL | CR | 9 | 0.11 | 0.14 |
| Waldenström's syndrome | Any | 10 | 0.13 | 0.06 |
| NHL | Any stage of mantle cell and low-grade; aggressive CR | 140 | 0.16 | 0.02 |
| ALL | CR1† | 28 | 0.17 | 0.04 |
| MM | CR | 38 | 0.19 | 0.06 |
| Standard risk | ||||
| CLL | No CR | 113 | 0.24 | 0.05 |
| CML | CP1 | 24 | 0.24 | 0.00 |
| MM | No CR | 179 | 0.32 | 0.17 |
| AML | CR‡ | 191 | 0.33 | 0.02 |
| MDS | RA/RARS | 30 | 0.35 | 0.00 |
| High risk | ||||
| NHL | Aggressive; no CR | 50 | 0.48 | 0.00 |
| AML | No CR; evolved from MDS | 98 | 0.65 | 0.04 |
| HL | After failed autologous HCT | 61 | 0.61 | 0.14 |
| MDS | RAEB; CMML; second | 62 | 0.65 | 0.04 |
| CML | CP2; AP; BC | 23 | 0.71 | 0.07 |
| ALL | ≥ CR2; no CR | 18 | 1.03 | — |
Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; AP, accelerated phase; BC, blast crisis; CLL, chronic lymphocytic leukemia; CML, chronic myelocytic leukemia; CMML, chronic myelomonocytic leukemia; CP, chronic phase; CR, complete remission; HCT, hematopoietic cell transplantation; HL, Hodgkin lymphoma; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasms; MM, multiple myeloma; NHL, non-Hodgkin lymphoma; RA, refractory anemia; RAEB, refractory anemia with excess blasts; RARS, refractory anemia with ring sideroblasts.
There were 243 patients in the low-risk group (53% related and 47% unrelated donors); 537 patients in the standard-risk group (58% related and 42% unrelated donors), and 312 patients in the high-risk group (54% related and 46% unrelated donors).
Before HCT, 14% of patients had minimal residual disease.
Before HCT, 13% of patients had minimal residual disease.
Fig 1.
Cumulative incidence rates of (A) relapse or progression, depending on risk group and (B) progression-free survival.
GVHD
Incidence.
The overall cumulative incidence rates of grades 2, 3, and 4 acute GVHD at 120 days were 37%, 9%, and 3%, respectively (Fig 2A); rates were lower for related than for unrelated recipients (32% v 44%, 8% v 11%, and 3% v 4%, respectively; P < .0001, both overall and grades 3 and 4 separately). NRM increased in direct relation to increasing grades of acute GVHD (Fig 2B). The 3-year cumulative incidence rates of chronic GVHD were comparable among related and unrelated recipients (49% and 50%, respectively). The cumulative incidence of any acute or chronic GVHD was 75% (71% for related recipients and 79% for unrelated recipients; P < .0001).
Fig 2.
Cumulative incidence rates of (A) acute graft-versus-host disease (GVHD) among 1,092 patients and (B) nonrelapse mortality in relation to acute GVHD grades.
NRM.
The day-100 cumulative incidence of NRM was 5% overall (4% for related recipients and 5% for unrelated recipients). The 1-year NRM was 15%. The 5-year NRM was 24% overall (20.6% for related recipients and 28.4% for unrelated recipients; Table 3). Most 5-year NRM was associated with GVHD (20.2%), either acute GVHD with (8.6%) or without (7.4%) chronic GVHD or de novo chronic GVHD (4.2%). Overall GVHD-associated mortality was 16.6% among related recipients and 24.9% among unrelated recipients. Forty patients (3.8%) died before 5 years without a history of GVHD, 18 with bacterial, fungal, or viral infections. Causes related to pretransplantation comorbidities predominated among the remaining non-GVHD deaths, including six cardiac and three pulmonary deaths.
Table 3.
5-Year NRM: Effects of Comorbidity* and GVHD
| GVHD | 5-Year NRM (%) |
||||
|---|---|---|---|---|---|
| All Patients | Patients With Related Donors |
Patients With Unrelated Donors |
|||
| HCT-CI 0-2 | HCT-CI ≥ 3 | HCT-CI 0-2 | HCT-CI ≥ 3 | ||
| Acute grade 0-1 | 3.8 | 2.7 | 6.1 | 2.5 | 4.2 |
| Acute grade 2-4 | |||||
| No chronic | 8.6† | 5.3 | 7.8 | 8.1 | 12.2 |
| With chronic | 7.4‡ | 4.6 | 6.4 | 11.6 | 13.3 |
| De novo chronic | 4.2 | 2.1 | 4.8 | 3.8 | 6.3 |
| Combined acute grade 2-4 and de novo chronic | 20.2 | 12 | 19 | 23.5 | 31.8 |
| Overall | 24 | 14.7 | 25.1 | 26 | 36 |
Abbreviations: GVHD, graft-versus-host disease; HCT-CI, hematopoietic cell transplantation comorbidity index; NRM, nonrelapse mortality.
HCT-CI scores had no association with incidence rates of acute or chronic GVHD.
NRM rates for acute grade 2 and grade 3 or 4 GVHD were 3.3% and 5.3%, respectively.
NRM rates for acute grade 2 and grade 3 or 4 GVHD were 5.2% and 2.2%, respectively.
Comorbidities.
Table 3 shows the combined effects of comorbidities and GVHD on cumulative 5-year NRM. Related recipients with HCT-CI scores of 0 to 2 experienced 14.7% NRM, of which 12% was NRM associated with acute or chronic GVHD; these figures increased to 25.1% and 19%, respectively, when scores were ≥ 3. Rates of NRM and NRM associated with acute or chronic GVHD were appreciably higher for unrelated recipients (26% and 23.5%, respectively, for those with scores of 0 to 2, and 36% and 31.8%, respectively, for those with scores of ≥ 3).
GHVD, relapse, NRM, and overall mortality.
Approximately half of the overall relapse incidence occurred in patients without a history of GVHD. During the first 2 years, their relapse rate was 0.50 (0.51 for related recipients and 0.48 for unrelated recipients) per patient year compared with 0.23 in patients with GVHD. Table 4 shows an association of acute GVHD with NRM but no significant GVT effects. Chronic GVHD was associated with significantly lower relapse or progression rates, but high associated NRM offset this benefit. These conclusions held true when patients with low-, standard-, and high-risk disease were analyzed separately (data not shown).
Table 4.
Time-Dependent Analysis of GVHD in Relation to Relapse, NRM, and Overall Mortality at 5 Years
| GVHD | Relapse or Progression |
NRM |
Overall Mortality |
||||||
|---|---|---|---|---|---|---|---|---|---|
| HR | 95% CI | P | HR | 95% CI | P | HR | 95% CI | P | |
| All patients (N = 1,092) | |||||||||
| No acute or chronic GVHD | 1.0 | 1.0 | 1.0 | ||||||
| Acute GVHD, grade 2* | 0.92 | 0.7 to 1.2 | .48 | 2.22 | 1.4 to 3.6 | .001 | 1.09 | 0.9 to 1.4 | .48 |
| Acute GVHD, grade 3 or 4* | 0.63 | 0.4 to 1.0 | .06 | 13.8 | 9.0 to 21.1 | < .0001 | 3.00 | 2.3 to 3.9 | < .0001 |
| Chronic after acute GVHD | 0.55 | 0.4 to 0.8 | .0002 | 4.83 | 3.2 to 7.8 | < .0001 | 1.10 | 0.9 to 1.4 | .42 |
| De novo chronic GVHD | 0.46 | 0.3 to 0.7 | < .001 | 3.52 | 2.2 to 5.8 | < .0001 | 0.88 | 0.7 to 1.1 | .32 |
| Related recipients with HCT-CI scores of 0-2 (n = 263) | |||||||||
| No acute or chronic GVHD | 1.0 | 1.0 | 1.0 | ||||||
| Acute GVHD, grade 2* | 0.75 | 0.4 to 1.3 | .31 | 1.62 | 0.5 to 5.8 | .46 | 0.91 | 0.5 to 1.7 | .78 |
| Acute GVHD, grade 3 or 4* | 1.25 | 0.6 to 2.7 | .57 | 15.4 | 5.5 to 42.8 | < .0001 | 3.63 | 2.0 to 6.6 | < .0001 |
| Chronic after acute GVHD | 0.43 | 0.2 to 0.8 | .008 | 3.46 | 1.2 to 9.6 | .02 | 0.95 | 0.6 to 1.5 | .82 |
| De novo chronic GVHD | 0.48 | 0.3 to 0.9 | .02 | 1.58 | 0.5 to 5.1 | .44 | 0.62 | 0.4 to 1.1 | .08 |
| Unrelated recipients with HCT-CI scores of 0-2 (n = 222) | |||||||||
| No acute or chronic GVHD | 1.0 | 1.0 | 1.0 | ||||||
| Acute GVHD, grade 2* | 0.90 | 0.5 to 1.5 | .70 | 1.57 | 0.5 to 5.1 | .46 | 0.78 | 0.4 to 1.4 | .38 |
| Acute GVHD, grade 3 or 4* | 0.57 | 0.2 to 1.6 | .29 | 13.1 | 4.4 to 36.7 | < .0001 | 1.93 | 1.0 to 3.6 | .04 |
| Chronic after acute GVHD | 0.60 | 0.3 to 1.2 | .14 | 4.14 | 1.5 to 12.4 | .009 | 1.09 | 0.6 to 1.8 | .75 |
| De novo chronic GVHD | 0.51 | 0.2 to 1.3 | .17 | 4.10 | 1.3 to 14.0 | .02 | 1.16 | 0.6 to 2.2 | .64 |
NOTE. The analysis was adjusted for relapse risk (low, standard, or high), comorbidity scores (0-2, ≥ 3, or missing), and GVHD prophylaxis (cyclosporine or tacrolimus).
Abbreviations: GVHD, graft-versus-host disease; HCT-CI, hematopoietic cell transplantation comorbidity index; HR, hazard ratio; NRM, nonrelapse mortality.
Before chronic GVHD.
When data among 263 patients with related grafts and low comorbidity scores were analyzed separately (Table 4), acute grade 3 or 4 GVHD was not associated with reduced relapse or progression but showed an association with high NRM and high overall mortality. Chronic GVHD arising from acute GVHD and de novo chronic GVHD remained associated with significantly reduced risk of relapse or progression, but this was offset by increased NRM, resulting in overall mortality hazard ratios (HRs) of 1.21 and 0.75, respectively. No significant GVT effects were seen with either acute or chronic GVHD among 222 unrelated recipients with low comorbidity scores (Table 4). Acute grade 3 or 4 GVHD was associated with increased risk of NRM. Results among all 481 unrelated recipients were comparable except for decreased relapse or progression with de novo chronic GVHD (HR, 0.50; 95% CI, 0.3 to 1.0; P = .04).
Risk factors for GVHD-related NRM.
A multivariate analysis using backward elimination showed HCT-CI scores of ≥ 3 to be the most significant risk factor for GVHD-related NRM (HR, 1.70; 95% CI, 1.3 to 2.3; P < .001), followed by unrelated transplantation (HR, 1.41; 95% CI, 1.1 to 1.9; P = .002) and CMV-positive status (HR, 1.35; 95% CI, 1.0 to 1.8; P = .04). Patients given cytoreductive autologous HCT for either MM or advanced NHL, 40 to 281 days before the allograft, had a reduced risk of GVHD-related NRM (HR, 0.55; 95% CI, 0.4 to 0.8; P = .003). Transplanted CD34 or CD3 cell doses, allele-level HLA class I mismatch, grafts from female donors into male recipients, the addition of fludarabine to TBI, the nature of the calcineurin inhibitor, and recipient age did not have significant effects on GVHD-related NRM, after adjustment for the four factors noted.
Survival
Five-year survival rates for patients in the low relapse risk group with HCT-CI scores of 0 to 2 and ≥ 3 were 60% and 42%, respectively. The figures were 50% and 35%, respectively, for patients in the standard relapse risk group and 30% and 25%, respectively, for patients in the high relapse risk group.
DISCUSSION
The current regimen avoids serious toxicities and lacks profound cytotoxic antitumor activities. Its main role is to enable sustained allogeneic engraftment, which was accomplished in 96% of patients, thereby setting in motion immunologic GVT effects.
The 5-year survival rate ranged from 25% to 60%, largely influenced by the following three factors: comorbid conditions, disease risk, and GVHD. GVHD in this older population with often considerable comorbidities was not more frequent than reported for younger patients given myeloablative conditioning.13,17 Powerful GVT effects were seen across all disease stages, except for advanced ALL. Although unrelated recipients experienced more GVHD than related recipients, the magnitude of GVT effects was comparable in the two patient groups. These effects presumably result from graft-versus-host reactions against minor histocompatibility antigens expressed on hematopoietic cells; however, natural killer cells may also be powerful contributors, especially for myeloid malignancies.18–22 Similar findings were reported in a Center for International Blood and Marrow Transplant Research analysis of patients with acute and chronic leukemias given a variety of myeloablative conditioning and postgrafting immunosuppression regimens.23 Seventy percent of relapse or progression occurred during year 1, and much of the remainder occurred during year 2. We hypothesize that early disease regrowth reflected a blunting of GVT effects as a result of early post-transplantation immune compromise and immunosuppression. Later, the immune system recovered, and immunosuppressive drugs were tapered or discontinued, permitting the donor immune cells to engage in GVT effects. Consistent with the hypothesis, relapse rates for all diseases, except MM and HL, were markedly reduced during years 3 to 5. The reasons for the partial escape of MM and HL cells from GVT effects are not clear.
Heightened GVT effects among current patients were associated with chronic GVHD but not with acute GVHD. Reducing or eliminating clinically significant acute GVHD would, therefore, not be expected to increase the risk of relapse. We have observed that CSP-treated related recipients whose donors were taking statins had no grade 3 or 4 acute GVHD and no apparent increase in relapse.24 If ongoing prospective trials confirm this retrospective observation, the 7.5% incidence of mortality associated with grade 3 or 4 acute GVHD could theoretically be eliminated, decreasing overall 5-year GVHD-related NRM from 20.2% to 12.7%. Most residual NRM (11.6%) would likely be related to chronic GVHD. In line with previous observations,4,17,25,26 chronic GVHD was associated with significantly decreased relapse risk compared with no GVHD, but this benefit was offset by an overall significantly higher NRM. As a result, patients with chronic GVHD preceded by acute GVHD experienced significantly increased mortality, whereas survival of patients with de novo chronic GVHD was comparable to that of patients without GVHD. Similarly, in subgroups of related and unrelated recipients with low comorbidity scores, neither de novo chronic GVHD nor chronic GVHD arising from acute GVHD conveyed significant survival advantages. These findings differed from early data published in 2005, which seemed to show a survival advantage for patients with chronic GVHD, attributable to associated GVT effects.25 The main difference is the reference groups. The current study calculated HRs relative to patients who had neither chronic nor acute GVHD, whereas the 2005 analysis was calculated relative to patients who had no chronic GVHD without regard for acute GVHD. Moreover, current patient numbers were much larger, and median follow-up time (5 years in the current study v 1.5 to 2 years in the 2005 analysis) was much longer, which more definitively captured late mortality both from relapse or progression and from chronic GVHD.
Taken together, current data suggest that effective prevention of chronic GVHD would decrease NRM but increase mortality related to relapse or progression. Although the resultant trading of one cause of death for another would not change overall mortality, effective prevention would avert the considerable morbidity associated with chronic GVHD and its treatment. Treatment of recipients with a statin medication offers a possible avenue for reducing the risk of chronic (but not acute) GVHD,27 although this approach might also increase the risk of relapse or progression.
Recent publications have addressed the use of reduced-intensity conditioning regimens for allogeneic HCT.27–34 As a rule, the regimens were more intense and thus less reliant on GVT effects than the current regimen. Most reports did not provide information on comorbidities. Five of the reports were retrospective National Marrow Donor Program/Center for International Blood and Marrow Transplant Research/European Bone Marrow Transplantation Group analyses,28–30,35 one was a multicenter French study,32 and three provided data from individual centers.31,33,34 In these studies, median follow-up after HCT was 3 years (range, 1 to 5 years). Across the studies, median event rates were 34% (range, 6% to 38%) for NRM and 43% (range, 22% to 65%) for relapse, with overall survival at 38% (range, 23% to 60%). One additional study showed that GVT effects were mainly associated with National Institutes of Health–defined chronic GVHD.26
Options for decreasing the risks of relapse and progression are limited. The use of more intense conditioning regimens would increase the risk of toxicity in elderly patients and those with comorbidities and, moreover, offer no benefit to the majority of current patients who did not experience relapse. Given these limitations, we hypothesize that improved cure rates could emerge from approaches that delay disease progression until the grafted immune system has recovered sufficiently to generate GVT effects. Well-tolerated targeted drugs or antibodies that are not curative on their own could set the stage for curative GVT effects. Candidate agents include antibodies to CD20 (NHL) and CD30 (HL),36 phosphoinositide 3′-kinase δ inhibitor CAL101 (CLL),37 proteasome inhibitors (MM), and FLT3 inhibitor AC220 (AML).38 As an example of such an approach, 3-year survival rates were 62% for patients with Ph1-positive ALL in first remission treated with imatinib for 1 year after HCT and 73% for the subgroup without minimal residual disease before HCT, which is impressively better than previous results without imatinib.39
We propose that GVT effects were present even without GVHD, assuming that relapse or progression would have been inevitable without HCT. This hypothesis is consistent with the notion that hematopoietic cells are targets for such reactions and echoes the International Bone Marrow Transplant Registry report describing less relapse in allogeneic recipients without GVHD compared with syngeneic recipients.4 Current patients who experienced chronic GVHD but not acute GVHD had significantly less relapse. Overall, the beneficial GVT effects associated with GVHD were offset by increased NRM. Major efforts should be directed toward methods that control progression of malignant disease early after HCT and more effectively prevent clinically significant GVHD.
Acknowledgment
We are grateful to all research nurses and data coordinators for implementation of protocols. We also thank our administrative staff for their assistance with manuscript preparation. We are grateful to the many physicians, nurses, physician assistants, nurse practitioners, pharmacists, and support staff who cared for our patients and to the patients who allowed us to care for them and who participated in our ongoing clinical research.
Appendix
Table A1.
Allogeneic Hematopoietic Cell Transplantation Protocols and Corresponding ClinicalTrials.Gov Registry Identifiers
| Protocol No. | ClinicalTrials.gov Identifier |
|---|---|
| 1209.00 | NCT00003145 |
| 1225.00 | NCT00003196 |
| 1383.00 | NCT00003954 |
| 1406.00 | NCT00005801 |
| 1409.00 | NCT00005803 |
| 1463.00 | NCT00005799 |
| 1533.00 | NCT00006251 |
| 1581.00 | NCT00036738 |
| 1591.00 | NCT00040846 |
| 1596.00 | NCT00014235 |
| 1641.00 | NCT00027820 |
| 1654.00 | NCT00045435 |
| 1668.00 | NCT00078858 |
| 1711.00 | NCT00060424 |
| 1732.00 | NCT00052546 |
| 1743.00 | NCT00054353 |
| 1813.00 | NCT00075478 |
| 1840.00 | NCT00104858 |
| 1898.00 | NCT00089011 |
| 1938.00 | NCT00105001 |
| 1959.00 | NCT00118352 |
| 2056.00 | NCT00397813 |
Footnotes
Listen to the podcast by Dr Apperley at www.jco.org/podcasts
Supported by Grants No. P01 HL036444, P01 CA078902, P01 CA018029, P30 CA015704, P30 CA023074, R01 HL108307, and R00 HL088021 from the National Institutes of Health, Bethesda, MD. Further support came from grants from the Leukemia/Lymphoma Society (Grant No. 7008-08), the Laura Landro Salomon Endowment Fund, the Danish Cancer Society, the Lundbeck Foundation, the Ricerca Finalizzata 2008-2009, and the Compagnia di San Paolo and Comitato Gigi Ghirotti.
The content of this article is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or its subsidiary institutes and centers.
Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTSOF INTEREST
The author(s) indicated no potential conflicts of interest.
AUTHOR CONTRIBUTIONS
Conception and design: Rainer Storb, Mohamed L. Sorror, David G. Maloney, Brenda M. Sandmaier
Financial support: Rainer Storb, Mohamed L. Sorror
Administrative support: Rainer Storb, H. Joachim Deeg
Provision of study materials or patients: Rainer Storb, Mohamed L. Sorror, Karl Blume, Dietger Niederwieser, Thomas R. Chauncey, Michael A. Pulsipher, Finn B. Petersen, Firoozeh Sahebi, Edward D. Agura, Parameswaran Hari, Benedetto Bruno, Peter A. McSweeney, Michael B. Maris, Richard T. Maziarz, Amelia A. Langston, Wolfgang Bethge, Lars Vindeløv, Georg-Nikolaus Franke, Ginna G. Laport, Andrew M. Yeager, Kai Hübel, H. Joachim Deeg, George E. Georges, Mary E.D. Flowers, Paul J. Martin, Marco Mielcarek, Ann E. Woolfrey, David G. Maloney, Brenda M. Sandmaier
Collection and assembly of data: Boglarka Gyurkocza, Mohamed L. Sorror, Karl Blume, Dietger Niederwieser, Thomas R. Chauncey, Michael A. Pulsipher, Finn B. Petersen, Firoozeh Sahebi, Edward D. Agura, Parameswaran Hari, Benedetto Bruno, Peter A. McSweeney, Michael B. Maris, Richard T. Maziarz, Amelia A. Langston, Wolfgang Bethge, Lars Vindeløv, Georg-Nikolaus Franke, Ginna G. Laport, Andrew M. Yeager, Kai Hübel, H. Joachim Deeg, George E. Georges, Mary E.D. Flowers, Paul J. Martin, Marco Mielcarek, Ann E. Woolfrey, David G. Maloney, Brenda M. Sandmaier
Data analysis and interpretation: Rainer Storb, Boglarka Gyurkocza, Barry E. Storer, Mohamed L. Sorror, Karl Blume, Ginna G. Laport, Marco Mielcarek, David G. Maloney, Brenda M. Sandmaier
Manuscript writing: All authors
Final approval of manuscript: All authors
REFERENCES
- 1.Barnes DWH, Corp MJ, Loutit JF, et al. Treatment of murine leukaemia with x-rays and homologous bone marrow: Preliminary communication. BMJ. 1956;2:626–627. doi: 10.1136/bmj.2.4993.626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mathe G, Amiel JL, Schwarzenberg L, et al. Successful allogeneic bone marrow transplantation in man: Chimerism, induced specific tolerance and possible anti-leukemia effects. Blood. 1965;25:179–196. [PubMed] [Google Scholar]
- 3.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. 1979;300:1068–1073. doi: 10.1056/NEJM197905103001902. [DOI] [PubMed] [Google Scholar]
- 4.Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood. 1990;75:555–562. [PubMed] [Google Scholar]
- 5.Apperley JF, Mauro FR, Goldman JM, et al. Bone marrow transplantation for chronic myeloid leukaemia in first chronic phase: Importance of a graft-versus-leukaemia effect. Br J Haematol. 1988;69:239–245. doi: 10.1111/j.1365-2141.1988.tb07628.x. [DOI] [PubMed] [Google Scholar]
- 6.Kolb HJ, Mittermüller J, Clemm C, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood. 1990;76:2462–2465. [PubMed] [Google Scholar]
- 7.Roddie C, Peggs KS. Donor lymphocyte infusion following allogeneic hematopoietic stem cell transplantation. Expert Opin Biol Ther. 2011;11:473–487. doi: 10.1517/14712598.2011.554811. [DOI] [PubMed] [Google Scholar]
- 8.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. 2001;97:3390–3400. doi: 10.1182/blood.v97.11.3390. [DOI] [PubMed] [Google Scholar]
- 9.Sorror ML, Maris MB, Storb R, et al. Hematopoietic cell transplantation (HCT)-specific comorbidity index: A new tool for risk assessment before allogeneic HCT. Blood. 2005;106:2912–2919. doi: 10.1182/blood-2005-05-2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Niederwieser D, Maris M, Shizuru JA, et al. Low-dose total body irradiation (TBI) and fludarabine followed by hematopoietic cell transplantation (HCT) from HLA-matched or mismatched unrelated donors and postgrafting immunosuppression with cyclosporine and mycophenolate mofetil (MMF) can induce durable complete chimerism and sustained remissions in patients with hematological diseases. Blood. 2003;101:1620–1629. doi: 10.1182/blood-2002-05-1340. [DOI] [PubMed] [Google Scholar]
- 11.Inamoto Y, Flowers ME, Appelbaum FR, et al. A retrospective comparison of tacrolimus versus cyclosporine with methotrexate for immunosuppression after allogeneic hematopoietic cell transplantation with mobilized blood cells. Biol Blood Marrow Transplant. 2011;17:1088–1092. doi: 10.1016/j.bbmt.2011.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2005;11:945–956. doi: 10.1016/j.bbmt.2005.09.004. [DOI] [PubMed] [Google Scholar]
- 13.Flowers ME, Inamoto Y, Carpenter PA, et al. Comparative analysis of risk factors for acute graft-versus-host disease and for chronic graft-versus-host disease according to National Institutes of Health consensus criteria. Blood. 2011;117:3214–3219. doi: 10.1182/blood-2010-08-302109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kahl C, Storer BE, Sandmaier BM, et al. Relapse risk among patients with malignant diseases given allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. Blood. 2007;110:2744–2748. doi: 10.1182/blood-2007-03-078592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53:457–481. [Google Scholar]
- 16.Gooley TA, Leisenring W, Crowley J, et al. Estimation of failure probabilities in the presence of competing risks: New representations of old estimators. Stat Med. 1999;18:695–706. doi: 10.1002/(sici)1097-0258(19990330)18:6<695::aid-sim60>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
- 17.Inamoto Y, Flowers ME, Lee SJ, et al. Influence of immunosuppressive treatment on risk of recurrent malignancy after allogeneic hematopoietic cell transplantation. Blood. 2011;118:456–463. doi: 10.1182/blood-2011-01-330217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ruggeri L, Capanni M, Casucci M, et al. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood. 1999;94:333–339. [PubMed] [Google Scholar]
- 19.Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–2100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
- 20.Miller JS, Cooley S, Parham P, et al. Missing KIR-ligands is associated with less relapse and increased graft versus host disease (GVHD) following unrelated donor allogeneic HCT. Blood. 2007;109:5058–5061. doi: 10.1182/blood-2007-01-065383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hsu KC, Keever-Taylor CA, Wilton A, et al. Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia (AML) predicted by KIR and HLA genotypes. Blood. 2005;105:4878–4884. doi: 10.1182/blood-2004-12-4825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Baron F, Petersdorf EW, Gooley T, et al. What is the role for donor natural killer cells after nonmyeolablative conditioning? Biol Blood Marrow Transplant. 2009;15:580–588. doi: 10.1016/j.bbmt.2009.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ringdén O, Pavletic SZ, Anasetti C, et al. The graft-versus-leukemia effect using matched unrelated donors is not superior to HLA-identical siblings for hematopoietic stem cell transplantation. Blood. 2009;113:3110–3118. doi: 10.1182/blood-2008-07-163212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Rotta M, Storer BE, Storb RF, et al. Donor statin treatment protects against severe acute graft-versus-host disease after related allogeneic hematopoietic cell transplantation. Blood. 2010;115:1288–1295. doi: 10.1182/blood-2009-08-240358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Baron F, Maris MB, Sandmaier BM, et al. Graft-versus-tumor effects after allogeneic hematopoietic cell transplantation with nonmyeloablative conditioning. J Clin Oncol. 2005;23:1993–2003. doi: 10.1200/JCO.2005.08.136. [DOI] [PubMed] [Google Scholar]
- 26.Thepot S, Zhou J, Perrot A, et al. The graft-versus-leukemia effect is mainly restricted to NIH-defined chronic graft-versus-host disease after reduced intensity conditioning before allogeneic stem cell transplantation. Leukemia. 2010;24:1852–1858. doi: 10.1038/leu.2010.187. [DOI] [PubMed] [Google Scholar]
- 27.Rotta M, Storer BE, Storb R, et al. Impact of recipient statin treatment on graft-versus-host disease after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2010;16:1463–1466. doi: 10.1016/j.bbmt.2010.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Pulsipher MA, Chitphakdithai P, Logan BR, et al. Donor, recipient, and transplant characteristics as risk factors after unrelated donor PBSC transplantation: Beneficial effects of higher CD34+ cell dose. Blood. 2009;114:2606–2616. doi: 10.1182/blood-2009-03-208355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Luger SM, Ringdén O, Zhang MJ, et al. Similar outcomes using myeloablative vs reduced-intensity allogeneic transplant preparative regimens for AML or MDS. Bone Marrow Transplant. 2012;47:203–211. doi: 10.1038/bmt.2011.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Giralt S, Logan B, Rizzo D, et al. Reduced-intensity conditioning for unrelated donor progenitor cell transplantation: Long-term follow-up of the first 285 reported to the National Marrow Donor Program. Biol Blood Marrow Transplant. 2007;13:844–852. doi: 10.1016/j.bbmt.2007.03.011. [DOI] [PubMed] [Google Scholar]
- 31.Blaise D, Farnault L, Faucher C, et al. Reduced-intensity conditioning with Fludarabin, oral Busulfan, and thymoglobulin allows long-term disease control and low transplant-related mortality in patients with hematological malignancies. Exp Hematol. 2010;38:1241–1250. doi: 10.1016/j.exphem.2010.08.012. [DOI] [PubMed] [Google Scholar]
- 32.Michallet M, Le QH, Mohty M, et al. Predictive factors for outcomes after reduced intensity conditioning hematopoietic stem cell transplantation for hematological malignancies: A 10-year retrospective analysis from the Societe Francaise de Greffe de Moelle et de Therapie Cellulaire. Exp Hematol. 2008;36:535–544. doi: 10.1016/j.exphem.2008.01.017. [DOI] [PubMed] [Google Scholar]
- 33.Warlick ED, Tomblyn M, Cao Q, et al. Reduced-intensity conditioning followed by related allografts in hematologic malignancies: Long-term outcomes most successful in indolent and aggressive non-Hodgkin lymphomas. Biol Blood Marrow Transplant. 2011;17:1025–1032. doi: 10.1016/j.bbmt.2010.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ho VT, Kim HT, Aldridge J, et al. Use of matched unrelated donors compared with matched related donors is associated with lower relapse and superior progression-free survival after reduced-intensity conditioning hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2011;17:1196–1204. doi: 10.1016/j.bbmt.2010.12.702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Belkacémi Y, Labopin M, Hennequin C, et al. Reduced-intensity conditioning regimen using low-dose total body irradiation before allogeneic transplant for hematologic malignancies: Experience from the European Group for Blood and Marrow Transplantation. Int J Radiat Oncol Biol Phys. 2007;67:544–551. doi: 10.1016/j.ijrobp.2006.08.049. [DOI] [PubMed] [Google Scholar]
- 36.Chen R, Gopal AK, Smith SG, et al. Results of a pivotal phase 2 study of brentuximab vedotin (SGN-35) in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2010;116:129. doi: 10.1182/blood-2014-08-595801. abstr 283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Furman RR, Byrd JC, Brown JR, et al. CAL-101, an isoform-selective inhibitor of phosphatidylinositol 3-kinase P110d, demonstrates clinical activity and pharmacodynamic effects in patients with relapsed or refractory chronic lymphocytic leukemia. Blood. 2010;116:31. abstr 55. [Google Scholar]
- 38.Cortes JE, Perl AE, Smith CC, et al. A phase II open-label AC220 monotherapy efficacy study in patients with refractory/relapsed FLT3-ITD positive acute myeloid leukemia: Updated interim results. Blood. 2011;118:2576. abstr. [Google Scholar]
- 39.Ram R, Storb R, Sandmaier BM, et al. Non-myeloablative conditioning with allogeneic hematopoietic cell transplantation for the treatment of high-risk acute lymphoblastic leukemia. Haematologica. 2011;96:1113–1120. doi: 10.3324/haematol.2011.040261. [DOI] [PMC free article] [PubMed] [Google Scholar]


