Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 188
Filtrer
1.
Cell Death Differ ; 31(4): 405-416, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38538744

RÉSUMÉ

BH3 mimetics, including the BCL2/BCLXL/BCLw inhibitor navitoclax and MCL1 inhibitors S64315 and tapotoclax, have undergone clinical testing for a variety of neoplasms. Because of toxicities, including thrombocytopenia after BCLXL inhibition as well as hematopoietic, hepatic and possible cardiac toxicities after MCL1 inhibition, there is substantial interest in finding agents that can safely sensitize neoplastic cells to these BH3 mimetics. Building on the observation that BH3 mimetic monotherapy induces AMP kinase (AMPK) activation in multiple acute leukemia cell lines, we report that the AMPK inhibitors (AMPKis) dorsomorphin and BAY-3827 sensitize these cells to navitoclax or MCL1 inhibitors. Cell fractionation and phosphoproteomic analyses suggest that sensitization by dorsomorphin involves dephosphorylation of the proapoptotic BCL2 family member BAD at Ser75 and Ser99, leading BAD to translocate to mitochondria and inhibit BCLXL. Consistent with these results, BAD knockout or mutation to BAD S75E/S99E abolishes the sensitizing effects of dorsomorphin. Conversely, dorsomorphin synergizes with navitoclax or the MCL1 inhibitor S63845 to induce cell death in primary acute leukemia samples ex vivo and increases the antitumor effects of navitoclax or S63845 in several xenograft models in vivo with little or no increase in toxicity in normal tissues. These results suggest that AMPK inhibition can sensitize acute leukemia to multiple BH3 mimetics, potentially allowing administration of lower doses while inducing similar antineoplastic effects.


Sujet(s)
AMP-Activated Protein Kinases , Dérivés de l'aniline , Protéine Mcl-1 , Pyrimidines , Sulfonamides , Protéine bcl-X , Humains , Animaux , Dérivés de l'aniline/pharmacologie , Sulfonamides/pharmacologie , AMP-Activated Protein Kinases/métabolisme , Souris , Protéine bcl-X/métabolisme , Protéine bcl-X/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Pyrimidines/pharmacologie , Protéine Mcl-1/métabolisme , Protéine Mcl-1/antagonistes et inhibiteurs , Pyrazoles/pharmacologie , Protéine Bad/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Mort cellulaire/effets des médicaments et des substances chimiques , Leucémies/traitement médicamenteux , Leucémies/anatomopathologie , Leucémies/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Fragments peptidiques/pharmacologie , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/antagonistes et inhibiteurs , Synergie des médicaments
2.
Leuk Res ; 139: 107481, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38484432

RÉSUMÉ

The BYOND study evaluated the efficacy and safety of bosutinib 500 mg once daily in patients with chronic myeloid leukemia (CML) resistant/intolerant to prior tyrosine kinase inhibitors (TKIs). These post-hoc analyses assessed the efficacy and safety of bosutinib by resistance or intolerance to prior TKIs (imatinib-resistant vs dasatinib/nilotinib-resistant vs TKI-intolerant), and cross-intolerance between bosutinib and prior TKIs (imatinib, dasatinib, nilotinib), in patients with Philadelphia chromosome-positive chronic phase CML. Data are reported after ≥3 years' follow-up. Of 156 patients with Philadelphia chromosome-positive chronic phase CML, 53 were imatinib-resistant, 29 dasatinib/nilotinib-resistant, and 74 intolerant to all prior TKIs; cumulative complete cytogenetic response rates at any time were 83.7%, 61.5%, and 86.8%, and cumulative major molecular response rates at any time were 72.9%, 40.7%, and 82.4%, respectively. Of 141, 95, and 79 patients who received prior imatinib, dasatinib, and nilotinib, 64 (45.4%), 71 (74.7%), and 60 (75.9%) discontinued the respective TKI due to intolerance; of these, 2 (3.1%), 5 (7.0%), and 0 had cross-intolerance with bosutinib. The response rates observed in TKI-resistant and TKI-intolerant patients, and low cross-intolerance between bosutinib and prior TKIs, further support bosutinib use for patients with Philadelphia chromosome-positive chronic phase CML resistant/intolerant to prior TKIs. TRIAL REGISTRATION: ClinicalTrials.gov: NCT02228382.


Sujet(s)
Dérivés de l'aniline , Antinéoplasiques , Leucémie myéloïde en phase chronique , Nitriles , Quinoléines , Humains , Mésilate d'imatinib/effets indésirables , Dasatinib/effets indésirables , Antinéoplasiques/effets indésirables , Chromosome Philadelphie , Inhibiteurs de protéines kinases/effets indésirables , Pyrimidines , Leucémie myéloïde en phase chronique/traitement médicamenteux ,
3.
J Natl Compr Canc Netw ; 22(1): 43-69, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38394770

RÉSUMÉ

Chronic myeloid leukemia (CML) is defined by the presence of Philadelphia chromosome resulting from a reciprocal translocation between chromosomes 9 and 22 [t9;22] that gives rise to a BCR::ABL1 fusion gene. CML occurs in 3 different phases (chronic, accelerated, and blast phase) and is usually diagnosed in the chronic phase in developed countries. Tyrosine kinase inhibitor (TKI) therapy is a highly effective treatment option for patients with chronic phase-CML. The primary goal of TKI therapy in patients with chronic phase-CML is to prevent disease progression to accelerated phase-CML or blast phase-CML. Discontinuation of TKI therapy with careful monitoring is feasible in selected patients. This manuscript discusses the recommendations outlined in the NCCN Guidelines for the diagnosis and management of patients with chronic phase-CML.


Sujet(s)
Leucémie myéloïde chronique BCR-ABL positive , Leucémie myéloïde en phase chronique , Humains , Crise blastique/induit chimiquement , Crise blastique/traitement médicamenteux , Crise blastique/génétique , Inhibiteurs de protéines kinases/effets indésirables , Leucémie myéloïde chronique BCR-ABL positive/diagnostic , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/génétique , Chromosome Philadelphie , Leucémie myéloïde en phase chronique/traitement médicamenteux , Protéines de fusion bcr-abl/génétique
7.
Leuk Lymphoma ; 64(4): 846-855, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36744656

RÉSUMÉ

The combination of venetoclax and hypomethylating agent (HMA/venetoclax) has emerged as a treatment option for patients with de novo acute myeloid leukemia (AML) who are unfit to receive intensive chemotherapy. In this single-center retrospective study, we evaluated clinical outcomes following treatment with HMA/venetoclax in 35 patients with advanced myeloproliferative neoplasms, myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes or AML with extramedullary disease. The composite complete remission (CR) rate (including confirmed/presumed complete cytogenetic response, acute leukemia response-complete, CR and CR with incomplete hematologic recovery) was 42.9% with median overall survival (OS) of 9.7 months. Complex karyotype was associated with inferior median OS (3.7 versus 12.2 months; p = 0.0002) and composite CR rate (22% versus 50.0%; p = 0.2444). Although SRSF2 mutations were associated with higher composite CR rate (80.0% versus 28.0%; p = 0.0082), this was not associated with longer median OS (10.9 versus 8.0 months; p = 0.2269). Future studies should include these patient subgroups.


Sujet(s)
Leucémie aigüe myéloïde , Syndromes myéloprolifératifs , Humains , Études rétrospectives , Composés hétérocycliques bicycliques/effets indésirables , Sulfonamides , Syndromes myéloprolifératifs/diagnostic , Syndromes myéloprolifératifs/traitement médicamenteux , Syndromes myéloprolifératifs/génétique , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Azacitidine/usage thérapeutique
8.
Transplant Cell Ther ; 29(3): 182.e1-182.e8, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36587740

RÉSUMÉ

Patients age ≥55 years with acute lymphoblastic leukemia (ALL) fare poorly with conventional chemotherapy, with a 5-year overall survival (OS) of ∼20%. Tyrosine kinase inhibitors and novel B cell-targeted therapies can improve outcomes, but rates of relapse and death in remission remain high. Allogeneic blood or marrow transplantation (alloBMT) provides an alternative consolidation strategy, and post-transplantation cyclophosphamide (PTCy) facilitates HLA-mismatched transplantations with low rates of nonrelapse mortality (NRM) and graft-versus-host disease (GVHD). The transplantation database at Johns Hopkins was queried for patients age ≥55 years who underwent alloBMT for ALL using PTCy. The database included 77 such patients. Most received reduced-intensity conditioning (RIC) (88.3%), were in first complete remission (CR1) (85.7%), and had B-lineage disease (90.9%). For the entire cohort, 5-year relapse-free survival (RFS) and overall survival (OS) were 46% (95% confidence interval [CI], 34% to 57%) and 49% (95% CI, 37% to 60%), respectively. Grade III-IV acute GVHD occurred in only 3% of patients, and chronic GVHD occurred in 13%. In multivariable analysis, myeloablative conditioning led to worse RFS (hazard ratio [HR], 4.65; P = .001), whereas transplantation in CR1 (HR, .30; P = .004) and transplantation for Philadelphia chromosome-positive (Ph+) ALL versus T-ALL (HR, .29; P = .03) were associated with improved RFS. Of the 54 patients who underwent RIC alloBMT in CR1 for B-ALL, the 5-year RFS and OS were 62% (95% CI, 47% to 74%) and 65% (95% CI, 51% to 77%), respectively, with a 5-year relapse incidence of 16% (95% CI, 7% to 27%) and an NRM of 24% (95% CI, 13% to 36%). RIC alloBMT with PTCy in CR1 represents a promising consolidation strategy for B-ALL patients age ≥55 years.


Sujet(s)
Maladie du greffon contre l'hôte , Transplantation de cellules souches hématopoïétiques , Leucémie-lymphome lymphoblastique à précurseurs B et T , Humains , Adulte d'âge moyen , Moelle osseuse , Cyclophosphamide/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Maladie du greffon contre l'hôte/traitement médicamenteux , Récidive , Maladie aigüe
9.
Clin Cancer Res ; 29(12): 2179-2183, 2023 06 13.
Article de Anglais | MEDLINE | ID: mdl-36547666

RÉSUMÉ

The FDA has an accelerated approval program for drugs that have been identified as promising treatments for serious conditions when the available data suggest that the benefits outweigh the foreseeable risks. All of the currently available treatment options for chronic myeloid leukemia (CML) initially went through the accelerated approval program. Here, a group of academic CML experts, patient panelists, and members from the FDA convened to discuss the utility of the accelerated approval program as it pertains to CML, and the utility of this program in future drug development in this disease. The results of that discussion are summarized here.


Sujet(s)
Agrément de médicaments , Leucémie myéloïde chronique BCR-ABL positive , États-Unis , Humains , Food and Drug Administration (USA) , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux
10.
Int J Hematol ; 117(3): 456-462, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36181657

RÉSUMÉ

Myeloproliferative neoplasms (MPN) are chronic clonal disorders characterized by overproduction of myeloid-lineage blood cells and potential risk of evolution to acute myeloid leukemia (AML). Chronic myeloid leukemia (CML) is distinct from other MPNs in that its pathophysiology stems from the BCR-ABL fusion protein of the Philadelphia chromosome (Ph +). Though there are known cases of Ph- and Ph + MPNs coexisting in a single patient, overall prevalence has never been quantified in a prospective cohort. Here, we review our center's MPN registry, which shows 0.6% of Ph- MPN patients later developed CML. This development occurred no less than 10 and up to 36 years after Ph- MPN diagnosis. This rate of chronic transformation exceeds what is expected, as the incidence of CML in the United States is 2 per 100,000 people-years. The probability of this CML case rate in an average-risk population is less than 0.001%, suggesting there are shared risk factors between Ph- and Ph + MPNs. We speculate that these risk factors may include exposures, genetic predispositions, or be inherent to disease biology. Abrupt-onset leukocytosis heralded post-MPN CML in all cases here and suggests this salient clinical feature should trigger hematologists to consider this diagnosis and perform appropriate testing.


Sujet(s)
Leucémie myéloïde chronique BCR-ABL positive , Syndromes myéloprolifératifs , Humains , Chromosome Philadelphie , Études prospectives , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/diagnostic , Leucémie myéloïde chronique BCR-ABL positive/épidémiologie , Leucémie myéloïde chronique BCR-ABL positive/génétique , Maladie chronique
11.
Leuk Lymphoma ; 63(8): 1942-1948, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35379077

RÉSUMÉ

There are currently no known predictors of myelodysplastic syndrome (MDS)/myeloproliferative overlap neoplasm (MPN) patients' response to hypomethylating agents (HMA). Forty-three patients with MDS/MPN who were treated with HMA during chronic phase and had next-generation sequencing using the established 63-genes panel were identified. Complete and partial remission and marrow response were assessed based on the MDS/MPN International Working Group response criteria. On univariate analysis, younger age, higher number of mutations, and mutations in SETBP1, RUNX1, or EZH2 were associated with no response. Multivariable analysis for modeling response were conducted via least absolute shrinkage and selection operator logistic regression approach, and showed that mutations in SETBP1, RUNX1, or EZH2 predict lack of HMA response. While limited by sample size, our findings suggest that genomic landscape can potentially identify MDS/MPN patients with lower likelihood of response to HMA.


Sujet(s)
Maladies myélodysplasiques-myéloprolifératives , Tumeurs , Sous-unité alpha 2 du facteur CBF/génétique , Génomique , Humains , Mutation , Maladies myélodysplasiques-myéloprolifératives/diagnostic , Maladies myélodysplasiques-myéloprolifératives/traitement médicamenteux , Maladies myélodysplasiques-myéloprolifératives/génétique
12.
Leuk Res Rep ; 17: 100304, 2022.
Article de Anglais | MEDLINE | ID: mdl-35371914

RÉSUMÉ

Acute promyelocytic leukemia (APL) is a rare acute leukemia generally considered curable with all-trans retinoic acid (ATRA) and arsenic trioxide (ATO). Some patients have co-morbidities that may limit the use of these agents and therefore impact curability. Adverse effects of ATO include life-threatening electrocardiographic abnormalities. ATO and its metabolites are partially excreted in the urine, and it is unclear to what extent ATO pharmacokinetics are impacted by hemodialysis. We present a patient on chronic hemodialysis successfully treated with ATO and ATRA for newly diagnosed APL. Complete molecular remission was achieved after induction and several drug-related toxicities were managed.

13.
Leuk Lymphoma ; 63(7): 1660-1668, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35244520

RÉSUMÉ

The phase 1b multicohort KEYNOTE-013 study assessed the safety and antitumor activity of pembrolizumab given at 10 mg/kg/day every 2 weeks for up to 2 years in hematologic malignancies, including myelodysplastic syndromes (MDS) refractory to a hypomethylating agent (HMA). Primary outcomes were safety and objective response rate per International Working Group 2006 criteria. By June 26, 2020, 28 patients were enrolled; median duration of follow-up was 5.6 months (range, 1-78), and 25 patients (89%) had died. Treatment-related adverse events occurred in 10 patients (36%), including 2 (7%) treatment-related discontinuations. No patient achieved complete or partial response. Five patients (19%) had bone marrow complete response, 12 (44%) stable disease, 10 (37%) progressive disease, 6 (22%) cytogenetic response, and 5 (19%) hematologic improvement. Median overall survival (OS) was 6.0 months (95% CI, 4-12); the overall 2-year OS rate was 17%. Pembrolizumab had manageable safety and clinical activity in patients with HMA-refractory MDS.This trial was registered at www.clinicaltrials.gov as #NCT01953692.


Sujet(s)
Anticorps monoclonaux humanisés , Syndromes myélodysplasiques , Anticorps monoclonaux humanisés/effets indésirables , Humains , Syndromes myélodysplasiques/diagnostic , Syndromes myélodysplasiques/traitement médicamenteux , Taux de survie
14.
Transplant Cell Ther ; 28(5): 259.e1-259.e11, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35158092

RÉSUMÉ

We describe outcomes after post-transplantation cyclophosphamide and nonmyeloablative conditioning-based allogeneic blood or marrow transplantation for myelofibrosis using matched or mismatched related or unrelated donors. The conditioning regimen consisted of fludarabine, cyclophosphamide, and total body irradiation. Forty-two patients were included, with a median age of 63 years, of whom 19% had Dynamic International Prognostic Scoring System (DIPSS)-plus intermediate-1 risk, 60% had intermediate-2 risk, and 21% had high-risk disease, and 60% had at least 1 high-risk somatic mutation. More than 90% of patients engrafted neutrophils, at a median of 19.5 days, and 7% experienced graft failure. At 1 year and 3 years, respectively, overall survival was 65% and 60%, relapse-free survival was 65% and 31%, relapse was 5% and 40%, and nonrelapse mortality was 30% and 30%. Acute graft-versus-host disease grade 3-4 was seen in 17% of patients at 1 year, and chronic graft-versus-host disease requiring systemic therapy in occurred in 12% patients. Spleen size ≥17 cm or prior splenectomy was associated with inferior relapse-free survival (hazard ratio [HR], 3.50; 95% confidence interval [CI], 1.18 to 10.37; P = .02) and higher relapse rate (subdistribution HR [SDHR] not calculable; P = .01). Age >60 years (SDHR, 0.26; 95% CI, 0.08 to 0.80, P = .02) and receipt of peripheral blood grafts (SDHR, 0.34; 95% CI, 0.11 to 0.99; P = .05) were associated with a lower risk of relapse. In our limited sample, the presence of a high-risk mutation was not statistically significantly associated with an inferior outcome, although ASXL1 was suggestive of inferior survival (SDHR, 2.36; 95% CI, 0.85 to 6.6; P = .09). Overall, this approach shows outcomes comparable those of to previously reported approaches and underscores the importance of spleen size in the evaluation of transplantation candidates.


Sujet(s)
Maladie du greffon contre l'hôte , Myélofibrose primitive , Moelle osseuse , Cyclophosphamide/usage thérapeutique , Maladie du greffon contre l'hôte/prévention et contrôle , Humains , Adulte d'âge moyen , Récidive tumorale locale/traitement médicamenteux , Myélofibrose primitive/thérapie
15.
Clin Cancer Res ; 28(13): 2753-2761, 2022 07 01.
Article de Anglais | MEDLINE | ID: mdl-35046058

RÉSUMÉ

PURPOSE: To evaluate efficacy and safety of venetoclax + azacitidine among treatment-naïve patients with IDH1/2-mutant (mut) acute myeloid leukemia (AML). PATIENTS AND METHODS: Data were pooled from patients enrolled in a phase III study (NCT02993523) that compared patients treated with venetoclax + azacitidine or placebo + azacitidine and a prior phase Ib study (NCT02203773) where patients were treated with venetoclax + azacitidine. Enrolled patients were ineligible for intensive therapy due to age ≥75 years and/or comorbidities. Patients on venetoclax + azacitidine received venetoclax 400 mg orally (days 1-28) and azacitidine (75 mg/m2; days 1-7/28-day cycle). RESULTS: In the biomarker-evaluable population, IDH1/2mut was detected in 81 (26%) and 28 (22%) patients in the venetoclax + azacitidine and azacitidine groups. Composite complete remission [CRc, complete remission (CR)+CR with incomplete hematologic recovery (CRi)] rates (venetoclax + azacitidine/azacitidine) among patients with IDH1/2mut were 79%/11%, median duration of remission (mDoR) was 29.5/9.5 months, and median overall survival (mOS) was 24.5/6.2 months. CRc rates among patients with IDH1/2 wild-type (WT) were 63%/31%, mDoR 17.5/10.3 months, and mOS 12.3/10.1 months. In patients with IDH1mut, CRc rates (venetoclax + azacitidine/azacitidine) were 66.7%/9.1% and mOS 15.2/2.2 months. In patients with IDH2mut, CRc rates were 86.0%/11.1% and mOS not reached (NR)/13.0 months. Patients with IDH1/2 WT AML treated with venetoclax + azacitidine with poor-risk cytogenetics had inferior outcomes compared with patients with IDH1/2mut, who had superior outcomes regardless of cytogenetic risk (mOS, IDH1/2mut: intermediate-risk, 24.5 months; poor-risk, NR; IDH1/2 WT: intermediate, 19.2 and poor, 7.4 months). There were no unexpected toxicities in the venetoclax + azacitidine group. CONCLUSIONS: Patients with IDH1/2mut who received venetoclax + azacitidine had high response rates, durable remissions, and significant OS; cytogenetic risk did not mitigate the favorable outcomes seen from this regimen for IDH1/2mut. See related commentary by Perl and Vyas, p. 2719.


Sujet(s)
Danse , Leucémie aigüe myéloïde , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Azacitidine/administration et posologie , Composés hétérocycliques bicycliques/usage thérapeutique , Humains , Isocitrate dehydrogenases/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/mortalité , Mutation , Sulfonamides
16.
Hematol Oncol ; 40(2): 269-279, 2022 Apr.
Article de Anglais | MEDLINE | ID: mdl-35043428

RÉSUMÉ

This study evaluated venetoclax population pharmacokinetics (popPK) in patients with treatment-naïve acute myeloid leukemia and assessed the relationship between venetoclax exposure and clinical response for venetoclax in combination with either a hypomethylating agent (HMA) or low-dose cytarabine (LDAC). A total of 771 patients who received venetoclax from 5 Phase 1-3 studies were included in the popPK model. Exposure-response analyses included data from 575 patients for venetoclax/placebo plus HMA and 279 patients for venetoclax/placebo plus LDAC. The popPK model successfully characterized venetoclax plasma concentrations over time and confirmed venetoclax exposure did not vary significantly with age, weight, sex, mild to moderate hepatic impairment, or mild to severe renal impairment. Asian patients had 67% higher mean relative bioavailability than non-Asian patients, however the range of exposures in Asian patients was similar to non-Asian patients. For all efficacy endpoints with both treatment combinations, efficacy was higher in the venetoclax treatment groups compared with the respective control arm of placebo plus azacitidine or LDAC. Within patients who received venetoclax, no significant exposure-efficacy relationships were identified for either treatment combination, indicating that the beneficial effects of venetoclax were already maximized in the dose ranges studied. There was no apparent effect of venetoclax exposure on treatment-emergent Grade ≥3 thrombocytopenia or infections for either combination. Rates of treatment-emergent Grade ≥3 neutropenia were higher in the venetoclax treatment arms compared with the respective control arms; however, within patients who received venetoclax, there was only a shallow relationship or no apparent relationship with venetoclax exposure for venetoclax plus HMA or LDAC, respectively. Along with the efficacy and safety data previously published, the exposure-response analyses support the venetoclax dose regimens of 400 mg once daily (QD) plus HMA and 600 mg QD plus LDAC in treatment-naïve AML patients who are ineligible for intensive chemotherapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Composés hétérocycliques bicycliques , Leucémie aigüe myéloïde , Sulfonamides , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Composés hétérocycliques bicycliques/effets indésirables , Composés hétérocycliques bicycliques/usage thérapeutique , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/étiologie , Sulfonamides/effets indésirables , Sulfonamides/usage thérapeutique
17.
Clin Lymphoma Myeloma Leuk ; 22(4): 260-269, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-34750086

RÉSUMÉ

INTRODUCTION: Mutations in the IDH1 or IDH2 genes are detected in approximately 20% of cases of acute myeloid leukemia (AML). Few studies have examined the impact of IDH mutations in AML on allogeneic bone marrow transplant (alloBMT) outcomes. PATIENTS AND METHODS: In this single center study, alloBMT outcomes for 61 patients with IDH-mutated (mIDH) AML were compared to those for 146 patients with IDH-wildtype (wtIDH) AML. RESULTS: Patients with mIDH AML had a 2-year overall survival (OS) of 85% (95% CI 76%-95%), 2-year relapse free survival (RFS) of 71% (95% CI 59%-85%), 1-year cumulative incidence of relapse (CIR) of 14% (95% CI 5%-23%) and a 1-year cumulative incidence of transplant related mortality (CITRM) of 3% (95% CI 0%-8%). Patients with wtIDH had a 2-year OS of 61% (95% CI 53%-70%), 2-year RFS of 58% (95% CI 50%-67%), 1-year CIR of 27% (95% CI 20%-35%), and a 1-year CITRM of 9% (95% CI 5%-14%). In a univariate analysis cox-proportional hazard model, mIDH was associated with significantly better OS (HR 0.52, 95% CI 0.29-0.96) and a trend toward better RFS (HR 0.60, 95% CI 0.35-1.01). After controlling for donor age, diagnosis, and ELN risk category, mIDH was associated with a nonsignificantly improved OS (HR 0.54, 95% CI 0.29-1.01) and RFS (HR 0.67, 95% CI 0.39-1.15). CONCLUSION: Among patients with mIDH AML, patients who received a peritransplant IDH inhibitor had improved OS (P = .03) compared to those who did not, but there was no detectable difference for RFS (P = .29).


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Leucémie aigüe myéloïde , Cyclophosphamide , Humains , Isocitrate dehydrogenases/génétique , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/thérapie , Mutation , Pronostic , Transplantation homologue
18.
Leuk Lymphoma ; 63(1): 64-73, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34702119

RÉSUMÉ

Real-world data are lacking to identify patients with secondary immunodeficiency (SID) who may benefit most from anti-infective interventions. This retrospective analysis used the IQVIA PharMetrics® Plus database to assess baseline characteristics associated with risk of severe infections post-SID diagnosis in patients with hematological malignancies. In 4066 patients included, the mean number of any and severe infections per patient in the one-year pre-SID diagnosis period was 9.5 and 0.7, respectively. Post-SID diagnosis, the mean annualized number of any and severe infections was 19.1 and 1.5, respectively. Receiver operating characteristic curve analysis identified a threshold (cutoff) of three bacterial infections at baseline as optimally predictive of severe infections post-SID diagnosis. Multivariate analysis indicated that hospitalizations, infections (≥3), or antibiotic use pre-SID diagnosis were predictive of severe infections post-SID diagnosis. Evaluation of these risk factors could inform clinical decisions regarding which patients may benefit from prophylactic anti-infective treatment, including immunoglobulin replacement if warranted.


Sujet(s)
Tumeurs hématologiques , Déficits immunitaires , Tumeurs hématologiques/complications , Tumeurs hématologiques/diagnostic , Tumeurs hématologiques/épidémiologie , Humains , Immunoglobulines , Déficits immunitaires/complications , Déficits immunitaires/diagnostic , Déficits immunitaires/épidémiologie , Études rétrospectives , Facteurs de risque
19.
J Oncol Pharm Pract ; 28(6): 1340-1349, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-34134554

RÉSUMÉ

Arsenic trioxide (ATO) and all-trans retinoic acid (ATRA) combination therapy yields high complete remission and disease-free survival rates in acute promyelocytic leukemia (APL). ATO is dosed on actual body weight and high ATO doses in overweight patients may contribute to increased toxicity. We performed a retrospective, two-center study comparing toxicities in patients who received the Lo-Coco et al ATRA/ATO regimen with capped ATO, ≤10 mg/dose, and non-capped ATO, >10 mg/dose. A total of 44 patients were included; 15 received doses ≤10 mg and 29 received >10 mg. During induction, there was no difference in the incidence of grade ≥3 hepatotoxicity, grade ≥3 QTc prolongation, neurotoxicity, and cardiac toxicity between groups. In consolidation, patients receiving >10 mg/dose experienced a greater incidence of neurotoxicity (66.7% vs 22.2%; p = 0.046). Capping doses saved $24634.37/patient and reduced waste of partially-used vials. At a median follow-up of 27 months, no disease relapses occurred in either group. This represents an opportunity to improve the safety profile of this highly effective regimen.


Sujet(s)
Composés de l'arsenic , Leucémie aiguë promyélocytaire , Protocoles de polychimiothérapie antinéoplasique , Trioxyde d'arsenic/effets indésirables , Composés de l'arsenic/effets indésirables , Humains , Leucémie aiguë promyélocytaire/traitement médicamenteux , Oxydes/effets indésirables , Études rétrospectives , Résultat thérapeutique , Trétinoïne/effets indésirables
20.
Blood Cancer Discov ; 2(6): 616-629, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34778801

RÉSUMÉ

Immune suppression, exhaustion, and senescence are frequently seen throughout disease progression in acute myeloid leukemia (AML). We conducted a phase II study of high-dose cytarabine followed by pembrolizumab 200 mg i.v. on day 14 to examine whether PD-1 inhibition improves clinical responses in relapsed/refractory (R/R) AML. Overall responders could receive pembrolizumab maintenance up to 2 years. Among 37 patients enrolled, the overall response rate, composite complete remission (CRc) rate (primary endpoint), and median overall survival (OS) were 46%, 38%, and 11.1 months, respectively. Patients with refractory/early relapse and those receiving treatment as first salvage had encouraging outcomes (median OS, 13.2 and 11.3 months, respectively). Grade ≥3 immune-related adverse events were rare (14%) and self-limiting. Patients who achieved CRc had a higher frequency of progenitor exhausted CD8+ T cells expressing TCF-1 in the bone marrow prior to treatment. A multifaceted correlative approach of genomic, transcriptomic, and immunophenotypic profiling offers insights on molecular correlates of response and resistance to pembrolizumab. SIGNIFICANCE: Immune-checkpoint blockade with pembrolizumab was tolerable and feasible after high-dose cytarabine in R/R AML, with encouraging clinical activity, particularly in refractory AML and those receiving treatment as first salvage regimen. Further study of pembrolizumab and other immune-checkpoint blockade strategies after cytotoxic chemotherapy is warranted in AML.See related commentary by Wei et al., p. 551. This article is highlighted in the In This Issue feature, p. 549.


Sujet(s)
Cytarabine , Leucémie aigüe myéloïde , Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Lymphocytes T CD8+ , Cytarabine/effets indésirables , Résistance aux médicaments antinéoplasiques , Humains , Leucémie aigüe myéloïde/traitement médicamenteux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...