10 results
Filters • 2
Sort By
Results Per Page
Filters
10 results
2
Download the following citations:
Email the following citations:
Print the following citations:
Editor's Choice
  • Sadowska-Klasa A
  • Zaucha JM
  • Labopin M
  • Bourhis JH
  • Blaise D
  • et al.
Bone Marrow Transplant. 2024 Mar;59(3):387-394 doi: 10.1038/s41409-023-02192-0.
POPULATION:

Adults with acute lymphoblastic leukaemia, reported to the EBMT registry (n=9720)

INTERVENTION:

A detailed analysis cohort who were transplanted for secondary acute lymphoblastic leukaemia (s-ALL, n=80)

COMPARISON:

Matched controls who were transplanted for de novo ALL (dn-ALL, n=80)

OUTCOME:

The estimated 2-year relapse incidence (RI) was 19.1% (95%CI: 11-28.9), leukemia-free survival (LFS) 52.1% (95%CI: 39.6-63.2), non-relapse mortality (NRM) 28.8% (95%CI: 18.4-40), GvHD-free, relapse-free survival (GRFS) 39.4% (95%CI: 27.8-50.7), and overall survival (OS) 60.8% (95%CI: 47.9-71.4), and did not differ between solid tumour and haematological disease patients. In a matched-pair analysis, there was no difference in RI, GRFS, NRM, LFS, or OS between s-ALL and dn-ALL except for a higher incidence of chronic GvHD (51.9% vs. 31.4%) in s-ALL.

Secondary acute lymphoblastic leukemia (s-ALL) comprises up to 10% of ALL patients. However, data regarding s-ALL outcomes is limited. To answer what is the role of allogeneic hematopoietic cell transplantation (HCT) in s-ALL, a matched-pair analysis in a 1:2 ratio was conducted to compare outcomes between s-ALL and de novo ALL (dn-ALL) patients reported between 2000-2021 to the European Society for Blood and Marrow Transplantation registry. Among 9720 ALL patients, 351 (3.6%) were s-ALL, of which 80 were in first complete remission (CR1) with a known precedent primary diagnosis 58.8% solid tumor (ST), 41.2% hematological diseases (HD). The estimated 2-year relapse incidence (RI) was 19.1% (95%CI: 11-28.9), leukemia-free survival (LFS) 52.1% (95%CI: 39.6-63.2), non-relapse mortality (NRM) 28.8% (95%CI: 18.4-40), GvHD-free, relapse-free survival (GRFS) 39.4% (95%CI: 27.8-50.7), and overall survival (OS) 60.8% (95%CI: 47.9-71.4), and did not differ between ST and HD patients. In a matched-pair analysis, there was no difference in RI, GRFS, NRM, LFS, or OS between s-ALL and dn-ALL except for a higher incidence of chronic GvHD (51.9% vs. 31.4%) in s-ALL. To conclude, patients with s-ALL who received HCT in CR1 have comparable outcomes to patients with dn-ALL.

Editor's Choice
  • Cabrero M
  • López-Corral L
  • Jarque I
  • de la Cruz-Vicente F
  • Pérez-López E
  • et al.
Bone Marrow Transplant. 2024 Mar;59(3):359-365 doi: 10.1038/s41409-023-02171-5.
POPULATION:

Adults with high-risk B-cell non-Hodgkin lymphoma with either chemorrefractory disease or who relapsed post-autologous transplant, from centres in Spain (n=33)

INTERVENTION:

Ofatumumab (OFA), a 2nd generation anti-CD20 moAb, added to a reduced intensity conditioning (RIC) regime

COMPARISON:

None

OUTCOME:

Acute GVHD affected 77% of patients (16% grade 3-4). Remarkably, GVHD achieved CR in 75% of patients after first-line treatment. Chronic GVHD, primarily mild or moderate, occurred in 54% of patients. NHL CR rate at day +100 was 81%. Relapses occurred in 7 patients after a median of 3 months. Causes of death were lymphoma progression (5), infections (10), and GVHD (2). At 24 months, progression-free and overall survival rates were 50.1 and 51.6% respectively.

Curative potential of allogeneic transplantation (AlloSCT) in high-risk non-Hodgkin lymphoma (NHL) could be enhanced by the integration of Ofatumumab (OFA), a 2nd generation anti-CD20 moAb, due to an antitumor effect and a role over graft-versus-host disease (GVHD). In this phase II trial (NCT01613300), we investigated safety and effectiveness of OFA-based reduced intensity conditioning (RIC). High-risk B-cell NHL patients with chemorrefractory disease or post-autologous SCT relapse were eligible. OFA was added to a standard RIC regimen. Primary endpoint was grade 3-4 aGVHD rate, while secondary endpoints included CR and survival rates. Thirty-three patients were included (median age 51; diffuse large B-cell:68%, HLA-identical donor: 74%). No grade >2 OFA toxicity was observed. Acute GVHD affected 77% of patients (16% grade 3-4). Remarkably, GVHD achieved CR in 75% of patients after first-line treatment. Chronic GVHD, primarily mild or moderate, occurred in 54% of patients. NHL CR rate at day +100 was 81%. Relapses occurred in 7 patients after a median of 3 months. Causes of death were lymphoma progression (5), infections (10), and GVHD (2). At 24 months, progression-free and overall survival rates were 50.1 and 51.6% respectively. OFA-RIC regimen is safe and effective, though acute GVHD remains a significant complication. However, data suggest that OFA could mitigate its severity.

Editor's Choice
  • Moukalled N
  • Labopin M
  • Versluis J
  • Socié G
  • Blaise D
  • et al.
Am J Hematol. 2024 Mar;99(3):360-369 doi: 10.1002/ajh.27187.
POPULATION:

Adults with NPM1-mutated de novo AML with known cytogenetic and FLT3-ITD status, reported to the EBMT registry (n=3275)

INTERVENTION:

Analysis of the impact of high-risk cytogenetics (CG) on allogeneic hematopoietic cell transplantation in the first complete remission.

COMPARISON:

None

OUTCOME:

Overall, 2377 (73%) patients had FLT3-ITD. On univariate analysis, only FLT3-ITD, minimal/measurable residual disease (MRD) positivity and CK, but not abnormal CG, affected posttransplant outcomes. On multivariable analysis, CK was associated with lower overall survival (OS) (hazard ratio [HR] 1.72). In the subgroup of 493 patients with aberrant CG, the 2-year leukemia-free survival (LFS) and OS were around 61% and 68%, respectively. On multivariable analysis for this subgroup, CK and MRD positivity were associated with increased risk of relapse (HR 1.7, p = .025; and 1.99) and worse LFS (HR 1.62, p = .018; and 1.64 respectively) while FLT3-ITD, MK, or other CG abnormalities had no significant effect. Importantly, CK negatively affected OS (HR 1.91). In the first complete remission transplant setting, CK was found as the only cytogenetic risk factor for worse outcomes in NPM1-mutated AML.

In the 2022 European LeukemiaNet classification, patients with nucleophosmin 1 (NPM1)-mutated acute myeloid leukemia (AML) were classified in the adverse-risk category in the presence of high-risk cytogenetics (CG). Nonetheless, the impact of various CG aberrations on posttransplant outcomes remains to be unraveled. This registry study analyzed adult patients with NPM1-mutated de novo AML who underwent their first allogeneic hematopoietic cell transplantation in the first complete remission from 2005 to 2021. A total of 3275 patients were identified, 2782 had normal karyotype, 493 had chromosomal aberrations including 160 with adverse-risk CG, 72 patients had complex karyotype (CK), and 66 monosomal karyotype (MK). Overall, 2377 (73%) patients had FLT3-ITD. On univariate analysis, only FLT3-ITD, minimal/measurable residual disease (MRD) positivity and CK, but not abnormal CG, affected posttransplant outcomes. On multivariable analysis, CK was associated with lower overall survival (OS) (hazard ratio [HR] 1.72, p = .009). In the subgroup of 493 patients with aberrant CG, the 2-year leukemia-free survival (LFS) and OS were around 61% and 68%, respectively. On multivariable analysis for this subgroup, CK and MRD positivity were associated with increased risk of relapse (HR 1.7, p = .025; and 1.99, p = .003 respectively) and worse LFS (HR 1.62, p = .018; and 1.64, p = .011 respectively) while FLT3-ITD, MK, or other CG abnormalities had no significant effect. Importantly, CK negatively affected OS (HR 1.91, p = .002). In the first complete remission transplant setting, CK was found as the only cytogenetic risk factor for worse outcomes in NPM1-mutated AML. Nevertheless, even for this subgroup, a significant proportion of patients can achieve long-term posttransplant survival.

Editor's Choice
  • Touzeau C
  • Perrot A
  • Hulin C
  • Manier S
  • Macro M
  • et al.
Blood. 2024 Feb 23; doi: 10.1182/blood.2023023597.
POPULATION:

Adults 65 years or younger with newly diagnosed myeloma from eleven centres in France (n=50)

INTERVENTION:

Daratumumab-carfilzomib-lenalidomide-dexamethasone (D-KRd) induction (6 cycles), autologous stem cell transplantation (ASCT), D-KRd consolidation (4 cycles), second ASCT, and daratumumab-lenalidomide maintenance for 2 years.

COMPARISON:

None

OUTCOME:

Median age was 57. Del(17p), t(4;14) and t(14;16) were found in 40%, 52% and 20% of patients respectively. At data cut-off, the study met the primary endpoint with 36 (72%) patients completing second transplant. Twenty patients (40%) discontinued the study due to stem-cell collection failure (n=8), disease progression (n=7), adverse event (n=4), consent withdrawal (n=1). Grade 3-4 Dara-KRd induction/consolidation related adverse events (>5% of patients) were neutropenia (39%), anemia (12%), thrombocytopenia (7%) and infection (6%). The overall response rate was 100% for patients completing second transplant (n=36), including 81% complete response. Pre-maintenance Minimal Residual Disease (MRD) negativity rate (NGS, 10-6) was 94%. After a median follow up of 33 months, the 30-month progression-free (PFS) and overall survival were 80% and 91%, respectively.

High-risk (HR) cytogenetics are associated with poor outcomes in newly diagnosed multiple myeloma (NDMM) and dedicated studies should address this difficult-to-treat population. The phase 2 study 2018-04 from the Intergroupe Francophone du Myelome evaluated feasibility of an intensive strategy with quadruplet induction and consolidation plus tandem transplant in HR transplant eligible (TE) NDMM (NCT03606577). HR cytogenetics were defined by the presence of del(17p), t(4;14) and/or t(14;16). Treatment consisted in daratumumab-carfilzomib-lenalidomide-dexamethasone (D-KRd) induction (6 cycles), autologous stem cell transplantation (ASCT), D-KRd consolidation (4 cycles), second ASCT, and daratumumab-lenalidomide maintenance for 2 years. The primary endpoint was feasibility. Fifty patients with previously untreated NDMM were included. Median age was 57. Del(17p), t(4;14) and t(14;16) were found in 40%, 52% and 20% of patients respectively. At data cut-off, the study met the primary endpoint with 36 (72%) patients completing second transplant. Twenty patients (40%) discontinued the study due to stem-cell collection failure (n=8), disease progression (n=7), adverse event (n=4), consent withdrawal (n=1). Grade 3-4 Dara-KRd induction/consolidation related adverse events (>5% of patients) were neutropenia (39%), anemia (12%), thrombocytopenia (7%) and infection (6%). The overall response rate was 100% for patients completing second transplant (n=36), including 81% complete response. Pre-maintenance Minimal Residual Disease (MRD) negativity rate (NGS, 10-6) was 94%. After a median follow up of 33 months, the 30-month progression-free (PFS) and overall survival were 80% and 91%, respectively. In conclusion, D-KRd with tandem transplant is feasible in high-risk TE NDMM patients and resulted in high rates of MRD negativity and PFS.

Editor's Choice
  • Meyran D
  • Arfeuille C
  • Chevret S
  • Neven Q
  • Caye-Eude A
  • et al.
POPULATION:

Consecutive children diagnosed with Juvenile myelomonocytic leukemia (JMML) in centres in France (n=119)

INTERVENTION:

First allogeneic HSCT between 2002 and 2021

COMPARISON:

None

OUTCOME:

The majority (97%) carried a RAS-pathway mutation, and 62% exhibited karyotypic alterations or additional mutations in SETBP1, ASXL1, JAK3 and/or the RAS pathway. Relapse was the primary cause of death, with a 5-year cumulative incidence of 24.6% (95%CI: 17.1-32.9). Toxic deaths occurred in 12 patients, resulting in treatment related mortality (TRM) of 9.0% (95%CI: 4.6-15.3). The 5-year overall (OS) and event-free survival were 73.6% (95%CI: 65.7-82.4) and 66.4% (95%CI: 58.2-75.8), respectively. Four independent adverse prognostic factors for OS were identified: age at diagnosis >2 years, time from diagnosis to HSCT >6 months, monocyte count at diagnosis >7.2x109/L, and the presence of additional genetic alterations. Based on these factors, we proposed a predictive classifier. Patients with three or more predictors (21% of the cohort) had a 5-year OS of 34.2%, whereas those with none (7%) had a 5-year OS of 100%.

Juvenile myelomonocytic leukemia (JMML) is an aggressive pediatric myeloproliferative neoplasm requiring hematopoietic stem cell transplantation (HSCT) in most cases. We retrospectively analyzed 119 JMML patients who underwent first allogeneic HSCT between 2002 and 2021. The majority (97%) carried a RAS-pathway mutation, and 62% exhibited karyotypic alterations or additional mutations in SETBP1, ASXL1, JAK3 and/or the RAS pathway. Relapse was the primary cause of death, with a 5-year cumulative incidence of 24.6% (95%CI: 17.1-32.9). Toxic deaths occurred in 12 patients, resulting in treatmentrelated mortality (TRM) of 9.0% (95%CI: 4.6-15.3). The 5-year overall (OS) and event-free survival were 73.6% (95%CI: 65.7-82.4) and 66.4% (95%CI: 58.2-75.8), respectively. Four independent adverse prognostic factors for OS were identified: age at diagnosis >2 years, time from diagnosis to HSCT >6 months, monocyte count at diagnosis >7.2x109/L, and the presence of additional genetic alterations. Based on these factors, we proposed a predictive classifier. Patients with three or more predictors (21% of the cohort) had a 5-year OS of 34.2%, whereas those with none (7%) had a 5-year OS of 100%. Our study demonstrates improved transplant outcomes compared to prior published data, which can be attributed to the synergistic impacts of a low TRM and a reduced yet still substantial relapse incidence. By integrating genetic information with clinical and hematological features, we have devised a predictive classifier. This classifier effectively identifies a subgroup of patients who are at a heightened risk of unfavorable post-transplant outcomes who would benefit novel therapeutic agents and post-transplant strategies.

Editor's Choice
  • Beelen DW
  • Iacobelli S
  • Koster L
  • Eikema DJ
  • van Biezen A
  • et al.
Bone Marrow Transplant. 2024 Feb 21; doi: 10.1038/s41409-024-02241-2.
POPULATION:

Adults aged 50-70 years with primary or secondary AML in complete remission or MDS, receiving a peripheral blood allogeneic transplant (n=1220)

INTERVENTION:

Participants in the MC-FludT.14/L trial who received fludarabine-treosulfan conditioning (FluTreo, n=252)

COMPARISON:

Propensity-score matched patients identified from the EBMT registry (n=968) who received fludarabine/melphalan (FluMel, n=338) or busulfan/cyclophosphamide (BuCy, n=630) regimens

OUTCOME:

In 1:1 propensity-score matched-paired analysis (PSA) of AML patients, there was no difference in 2-year-relapse-incidence after FluTreo compared with either FluMel (n=110) or BuCy (n=78). However, 2-year-non-relapse-mortality (NRM) was lower compared with FluMel and BuCy. Consequently, 2-year-overall-survival (OS) after FluTreo was higher compared with FluMel and BuCy. For MDS patients, no endpoint differences between FluTreo and FluMel (n=30) were evident, whereas 2-year-OS after FluTreo was higher compared with BuCy (n=25) due to lower 2-year-NRM. Multivariate sensitivity analysis confirmed all significant results of PSA.

A randomized study (acronym: MC-FludT.14/L Trial II) demonstrated that fludarabine plus treosulfan (30 g/m²) was an effective and well tolerated conditioning regimen for allogeneic hematopoietic cell transplantation (allo-HCT) in older patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). To further evaluate this regimen, all 252 study patients aged 50 to 70 years were compared with similar patients, who underwent allo-HCT after fludarabine/melphalan (140 mg/m²) (FluMel) or busulfan (12.8 mg/kg)/cyclophosphamide (120 mg/kg) (BuCy) regimens and whose data was provided by the European Society for Blood and Marrow Transplantation registry. In 1:1 propensity-score matched-paired analysis (PSA) of AML patients, there was no difference in 2-year-relapse-incidence after FluTreo compared with either FluMel (n = 110, p = 0.28) or BuCy (n = 78, p = 0.98). However, 2-year-non-relapse-mortality (NRM) was lower compared with FluMel (p = 0.019) and BuCy (p < 0.001). Consequently, 2-year-overall-survival (OS) after FluTreo was higher compared with FluMel (p = 0.04) and BuCy (p < 0.001). For MDS patients, no endpoint differences between FluTreo and FluMel (n = 30) were evident, whereas 2-year-OS after FluTreo was higher compared with BuCy (n = 25, p = 0.01) due to lower 2-year-NRM. Multivariate sensitivity analysis confirmed all significant results of PSA. Consequently, FluTreo (30 g/m²) seems to retain efficacy compared with FluMel and BuCy, but is better tolerated by older patients.

Editor's Choice
  • Fein JA
  • Shouval R
  • Krieger E
  • Spellman SR
  • Wang T
  • et al.
Blood Adv. 2024 Feb 13;8(3):581-590 doi: 10.1182/bloodadvances.2023011622.
POPULATION:

Adults with acute myeloid leukaemia who underwent matched unrelated donor (MUD) transplantation in complete remission, identified from the CIBMTR registry (n=2025)

INTERVENTION:

Evaluation of individual donor-KIR/recipient-HLA interactions for HCT outcomes and empirically evaluated prevalent KIR genotypes for clinical benefit.

COMPARISON:

Independent validation cohort of 796 AML transplants from the German transplantation registry (n=796)

OUTCOME:

Only the donor-2DL2+/recipient-HLA-C1+ pair was associated with reduced relapse (hazard ratio 0.79 [95% confidence interval: 0.67, 0.93]) compared with donor-2DL2-/recipient-HLA-C1+. However, no association were found when comparing HLA-C groups among KIR-2DL2+-graft recipients. We identified nine prevalent donor KIR genotypes in our cohort and screened them for association with relapse risk. Genotype (G)5 in all recipients and G3 in Bw4+recipients were associated with decreased relapse risk (HR 0.52 [0.35, 0.78]; 0.32 [0.14, 0.72], respectively) and G2 (HR 1.63 [1.15, 2.29]) with increased relapse risk C1-homozygous recipients, compared to patients with the same ligand.

In acute myeloid leukemia (AML), donor natural killer cell killer immunoglobulin-like receptors (KIR) and recipient HLA interactions may contribute to the graft-versus-leukemia effect of allogeneic hematopoietic cell transplantation (HCT). Analyses of individual KIR/HLA interactions, however, have yielded conflicting findings, and their importance in the HLA-matched unrelated donor (MUD) setting remains controversial. We systematically studied outcomes of individual donor-KIR/recipient-HLA interactions for HCT outcomes and empirically evaluated prevalent KIR genotypes for clinical benefit. Adult patients with AML (n = 2025) who received HCT with MUD grafts in complete remission reported to the Center for International Blood and Marrow Transplantation were evaluated. Only the donor-2DL2+/recipient-HLA-C1+ pair was associated with reduced relapse (hazard ratio [HR], 0.79; 95% confidence interval [CI], 0.67-0.93; P = .006) compared with donor-2DL2-/recipient-HLA-C1+ pair. However, no association was found when comparing HLA-C groups among KIR-2DL2+-graft recipients. We identified 9 prevalent donor KIR genotypes in our cohort and screened them for association with relapse risk. Genotype 5 (G5) in all recipients and G3 in Bw4+ recipients were associated with decreased relapse risk (HR, 0.52; 95% CI, 0.35-0.78; P = .002; and HR, 0.32; 95% CI, 0.14-0.72; P = .006; respectively) and G2 (HR 1.63, 95% CI, 1.15-2.29; P = .005) with increased relapse risk in C1-homozygous recipients, compared with other patients with the same ligand. However, we could not validate these findings in an external data set of 796 AML transplants from the German transplantation registry. Neither a systematic evaluation of known HLA-KIR interactions nor an empiric assessment of prevalent KIR genotypes demonstrated clinically actionable associations; therefore, these data do not support these KIR-driven strategies for MUD selection in AML.

Editor's Choice
  • Al Hadidi S
  • Ababneh OE
  • Schinke CD
  • Thanendrarajan S
  • Bailey C
  • et al.
Blood Adv. 2024 Feb 13;8(3):703-707 doi: 10.1182/bloodadvances.2023011601.
POPULATION:

Adults from a single centre in USA with newly diagnosed multiple myeloma, enrolled in the Total Therapy (TT) IIIB trial between 2006 and 2008, (n=177)

INTERVENTION:

Bortezomib incorporated into tandem melphalan-based hematopoietic stem cell transplantation with dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, and etoposide for induction/consolidation and bortezomib, lenalidomide, and dexamethasone (VRD) for maintenance. Three years of VRD maintenance (TT IIIB, n=177)

COMPARISON:

Comparison cohort received one year of bortezomib, thalidomide, and dexamethasone followed by two years of thalidomide and dexamethasone (TD) maintenance (TT IIIA group)

OUTCOME:

15-year progression free survival (PFS) in TTIIIB was 27.9%. Median PFS was better in Gene expression profile (GEP)-defined low-risk patients at 7.8 years and in international stage system stage I patients at 8.7 years. Overall, median overall survival (OS) was 9.1 years, and 15-year OS was 35.9%. GEP-defined low-risk patients' median OS was 11.2 years, and that of GEP-defined high-risk patients was 2.8 years. There was no difference in OS between TT IIIB and TT IIIA.

The total therapy (TT) IIIB phase 2 study incorporated bortezomib into tandem melphalan-based hematopoietic stem cell transplantation with dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, and etoposide for induction/consolidation and bortezomib, lenalidomide, and dexamethasone (VRD) for maintenance in patients with newly diagnosed multiple myeloma (MM). This updated analysis presents a 15.4-year median follow-up. Of 177 patients, 21% patients had gene expression profile (GEP)-defined high-risk MM. 15-year progression free survival (PFS) was 27.9%. Median PFS was better in GEP-defined low-risk patients at 7.8 years and in International Staging System stage 1 patients at 8.7 years. Overall, median OS was 9.1 years, and 15-year overall survival (OS) was 35.9%. GEP-defined low-risk patients' median OS was 11.2 years, and that of GEP-defined high-risk patients was 2.8 years. There was no difference in OS between TT IIIB and TT IIIA. This study includes the longest follow-up of patients treated with maintenance VRD reported to date. In patients with GEP-defined low-risk, nearly half and one-third of patients without ongoing treatment showed no signs of progression at 10 and 15 years, respectively. One-third of patients survived more than 15 years, but 3 years of VRD maintenance did not improve outcomes for patients with GEP-defined high-risk MM. The study was registered on www.clinicaltrials.gov as #NCT00572169.

Editor's Choice
  • Pasvolsky O
  • Saliba RM
  • Popat UR
  • Alousi A
  • Mehta R
  • et al.
Clin Lymphoma Myeloma Leuk. 2024 Feb 3; doi: 10.1016/j.clml.2024.01.012.
POPULATION:

Adults with FLT3-negative acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS)

INTERVENTION:

Post-transplant 5-azacitidine (AZA) maintenance off clinical trial (n = 93).

COMPARISON:

Contemporaneous AML/MDS patients who did not receive any maintenance (n = 357)

OUTCOME:

The AZA and control groups had comparable patient and disease characteristics except for older age (median: 61 vs. 57 years) and lower hematopoietic comorbidity index (median: 2 vs. 3) in the AZA group. The 3-year cumulative incidence of progression in the AZA and control groups was 29% vs. 33%. The protective effect of AZA on progression was limited to patients with high-risk AML/MDS (HR = 0.4, 95% CI = 0.2-0.8). This led to improved progression-free survival both in high-risk AML and MDS patients with maintenance (HR = 0.2, 95% CI = 0.1-0.6, and HR = 0.4, 95% CI = 0.2-0.9).

BACKGROUND:

Maintenance after allogeneic hematopoietic cell transplantation (alloHCT) with hypomethylating agents has yielded conflicting results.

MATERIALS AND METHODS:

We conducted a single center retrospective matched-control analysis with the study group (5-azacitidine [AZA] group) including adults with FLT3-negative acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) who received post-transplant AZA maintenance off clinical trial (n = 93). A matched control group was comprised of contemporaneous AML/MDS patients who did not receive any maintenance (n = 357). Primary endpoint was disease progression.

RESULTS:

The AZA and control groups had comparable patient and disease characteristics except for older age (median: 61 vs. 57 years, P = .01) and lower hematopoietic comorbidity index (median: 2 vs. 3, P = .04) in the AZA group. The 3-year cumulative incidence of progression in the AZA and control groups was 29% vs. 33% (P = .09). The protective effect of AZA on progression was limited to patients with high-risk AML/MDS (HR = 0.4, 95% CI = 0.2-0.8, P = .009). This led to improved progression-free survival both in high-risk AML and MDS patients with maintenance (HR = 0.2, 95% CI = 0.1-0.6, P = .004 and HR = 0.4, 95% CI = 0.2-0.9, P = .04).

CONCLUSION:

AZA maintenance was associated with a lower progression rate in patients with high-risk FLT3-negative AML or MDS, and AZA maintenance should be considered for post-alloHCT maintenance in this subset.

Editor's Choice
  • Pasvolsky O
  • Ghanem S
  • Milton DR
  • Rauf M
  • Tanner MR
  • et al.
Blood Cancer J. 2024 Jan 10;14(1):4 doi: 10.1038/s41408-023-00973-w.
POPULATION:

Adults with newly-diagnosed myeloma (NDMM) patients with 1q21 gain/amplification (3 or >/=4 copies of 1q, respectively) that received autoSCT between from a single centre in USA (n=213)

INTERVENTION:

Assess the prognostic impact of additional copies of chromosome 1q (1q+) on autologous transplantation outcomes

COMPARISON:

None

OUTCOME:

At day100 post-autoSCT and at best post-transplant response, 78% and 87% of patients achieved very good partial response or better (>/=VGPR) and 38% and 50% achieved MRD-negative >/=VGPR, respectively. Median PFS and OS for the entire cohort were 35.5 months and 81.4 months, respectively. On multivariable assessment for PFS, MRD negative >/=VGPR before autoSCT (HR 0.52) was associated with superior PFS, whereas 1q amplification was associated with inferior PFS (2.03). On multivariate analysis for OS, achieving MRD negative >/=VGPR at best post-transplant response was associated with superior survival (0.29), whereas R-ISS III and concomitant del17p or t(4:14) were associated with inferior survival (6.95, 2.33, and 3.00, respectively).

The prognostic impact of additional copies of chromosome 1q (1q + ) on outcomes of newly-diagnosed multiple myeloma (NDMM) patients undergoing autologous transplantation (autoSCT) is unclear. We conducted a retrospective single-center analysis of NDMM patients with 1q21 gain/amplification (3 or ≥4 copies of 1q, respectively) that received autoSCT between 2008-2018. 213 patients were included (79% 1q gain; 21% 1q amplification). The most commonly used induction regimen was bortezomib, lenalidomide, and dexamethasone (41%). At day100 post-autoSCT and at best post-transplant response, 78% and 87% of patients achieved ≥VGPR, and 38% and 50% achieved MRD-negative ≥VGPR, respectively. Median PFS and OS for the entire cohort were 35.5 months and 81.4 months, respectively. On multivariable assessment for PFS, MRD negative ≥VGPR before autoSCT (HR 0.52, p = 0.013) was associated with superior PFS, whereas 1q amplification was associated with inferior PFS (2.03, p = 0.003). On multivariate analysis for OS, achieving MRD negative ≥VGPR at best post-transplant response was associated with superior survival (0.29, p < 0.001), whereas R-ISS III and concomitant del17p or t(4:14) were associated with inferior survival (6.95, p = 0.030, 2.33, p = 0.023 and 3.00, p = 0.047, respectively). In conclusion, patients with 1q+ NDMM, especially 1q amplification, have inferior survival outcomes compared to standard-risk disease after upfront autoSCT, though outcomes are better than other high-risk cytogenetic abnormalities.