Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
1.
Front Immunol ; 13: 970287, 2022.
Article de Anglais | MEDLINE | ID: mdl-36466858

RÉSUMÉ

Severe respiratory viral infections, including SARS-CoV-2, have resulted in high mortality rates despite corticosteroids and other immunomodulatory therapies. Despite recognition of the pathogenic role of neutrophils, in-depth analyses of this cell population have been limited, due to technical challenges of working with neutrophils. We undertook an unbiased, detailed analysis of neutrophil responses in adult patients with COVID-19 and healthy controls, to determine whether distinct neutrophil phenotypes could be identified during infections compared to the healthy state. Single-cell RNA sequencing analysis of peripheral blood neutrophils from hospitalized patients with mild or severe COVID-19 disease and healthy controls revealed distinct mature neutrophil subpopulations, with relative proportions linked to disease severity. Disruption of predicted cell-cell interactions, activated oxidative phosphorylation genes, and downregulated antiviral and host defense pathway genes were observed in neutrophils obtained during severe compared to mild infections. Our findings suggest that during severe infections, there is a loss of normal regulatory neutrophil phenotypes seen in healthy subjects, coupled with the dropout of appropriate cellular interactions. Given that neutrophils are the most abundant circulating leukocytes with highly pathogenic potential, current immunotherapies for severe infections may be optimized by determining whether they aid in restoring an appropriate balance of neutrophil subpopulations.


Sujet(s)
COVID-19 , Humains , Granulocytes neutrophiles , SARS-CoV-2 , Acuité des besoins du patient , Antiviraux
2.
J Cell Mol Med ; 25(24): 11039-11052, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34791807

RÉSUMÉ

Acute myeloid leukaemia (AML) is an aggressive form of blood cancer that carries a dismal prognosis. Several studies suggest that the poor outcome is due to a small fraction of leukaemic cells that elude treatment and survive in specialised, oxygen (O2 )-deprived niches of the bone marrow. Although several AML drug targets such as FLT3, IDH1/2 and CD33 have been established in recent years, survival rates remain unsatisfactory, which indicates that other, yet unrecognized, mechanisms influence the ability of AML cells to escape cell death and to proliferate in hypoxic environments. Our data illustrates that Carbonic Anhydrases IX and XII (CA IX/XII) are critical for leukaemic cell survival in the O2 -deprived milieu. CA IX and XII function as transmembrane proteins that mediate intracellular pH under low O2 conditions. Because maintaining a neutral pH represents a key survival mechanism for tumour cells in O2 -deprived settings, we sought to elucidate the role of dual CA IX/XII inhibition as a novel strategy to eliminate AML cells under hypoxic conditions. Our findings demonstrate that the dual CA IX/XII inhibitor FC531 may prove to be of value as an adjunct to chemotherapy for the treatment of AML.


Sujet(s)
Antinéoplasiques/pharmacologie , Carbonic anhydrase IX/antagonistes et inhibiteurs , Inhibiteurs de l'anhydrase carbonique/pharmacologie , Carbonic anhydrases/métabolisme , Hypoxie tumorale/effets des médicaments et des substances chimiques , Adulte , Sujet âgé , Animaux , Antigènes néoplasiques/génétique , Carbonic anhydrase IX/génétique , Carbonic anhydrases/génétique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Modèles animaux de maladie humaine , Synergie des médicaments , Femelle , Duplication de gène , Expression des gènes , Humains , Concentration en ions d'hydrogène , Immunohistochimie , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/étiologie , Leucémie aigüe myéloïde/métabolisme , Mâle , Adulte d'âge moyen , Hypoxie tumorale/génétique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Jeune adulte , Tyrosine kinase-3 de type fms/génétique
3.
Blood Adv ; 5(23): 5047-5056, 2021 12 14.
Article de Anglais | MEDLINE | ID: mdl-34607341

RÉSUMÉ

A potent graft-versus-leukemia (GVL) response is crucial in preventing relapse, the major impediment to successful allogeneic hematopoietic cell transplantation (HCT). In preclinical studies, type 1 interferon (IFN-α) enhanced cross-presentation of leukemia-specific antigens by CD8α dendritic cells (DCs) and amplified GVL. This observation was translated into a proof-of-concept phase 1/2 clinical trial with long-acting IFN-α (pegylated IFN-α [pegIFNα]) in patients undergoing HCT for high-risk acute myeloid leukemia (AML). Patients with treatment-resistant AML not in remission or those with poor-risk leukemia were administered 4 dosages of pegIFNα every 14 days beginning at day -1 before HCT. Dose selection was established by adaptive design that continuously assessed the probability of dose-limiting toxicities throughout the trial. Efficacy was evaluated by determining the 6-month incidence of relapse at the maximum tolerated dose (MTD). Thirty-six patients (median age, 60 years) received pegIFNα treatment. Grade 3 or greater severe adverse events occurred in 25% of patients, establishing 180 µg as the MTD. In phase 2, the incidence of relapse was 39% at 6 months, which was sustained through 1-year post-HCT. The incidence of transplant-related mortality was 13%, and severe grade III-IV acute graft-versus-host disease (GVHD) occurred in 11%. Paired blood samples from donors and recipients after HCT revealed elevated levels of type 1 IFN with cellular response, the persistence of cross-presenting DCs, and circulating leukemia antigen-specific T cells. These data suggest that prophylactic administration of pegIFNα is feasible in the peri-HCT period. In high-risk AML, increased toxicity was not observed with preliminary evidence for reduction in leukemia relapse after HCT. This trial was registered at www.clinicaltrials.gov as #NCT02328755.


Sujet(s)
Maladie du greffon contre l'hôte , Leucémie aigüe myéloïde , Maladie du greffon contre l'hôte/étiologie , Maladie du greffon contre l'hôte/prévention et contrôle , Humains , Interféron alpha/usage thérapeutique , Leucémie aigüe myéloïde/thérapie , Adulte d'âge moyen , Récidive , Transplantation homologue
4.
Oncogene ; 39(14): 3015-3027, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32060420

RÉSUMÉ

TP53 mutation in acute myeloid leukemia (AML) is associated with poor prognosis. Since no targeted therapy is available to restore p53 function, it is of great interest to test whether other pathways activated by TP53 mutations can be therapeutically targeted. Here, we showed HIF-1α target genes are enriched in TP53-mutated versus TP53-wild-type AML. To determine the role of this activation, we tested efficacy of HIF-1α inhibitor echinomycin in TP53-mutated AML samples in vitro and in vivo. Echinomycin was broadly effective against a panel of primary AML blast cells, with low nanomolar IC50s and, based on colony-forming unit assay, was tenfold more effective in eliminating AML stem cells. Echinomycin selectively eliminated CD34+CD38- AML cells. To test the therapeutic efficacy of echinomycin, we established a xenograft model of TP53-mutated AML. Echinomycin was broadly effective against xenografts from multiple AML samples in vivo, and more effective than cytarabine + daunorubicin chemotherapy. Importantly, while cytarabine + daunorubicin enriched for AML stem cells, echinomycin nearly eliminated this population. Using TP53-mutated AML cell line THP1 and patient-derived AML cells, we tested a new echinomycin formulation with longer half-life and significantly improved therapeutic effect. Our data suggest a novel approach to treat AML with TP53 mutations.


Sujet(s)
Échinomycine/pharmacologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Mutation/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Lignée cellulaire tumorale , Femelle , Humains , Leucémie aigüe myéloïde/génétique , Mâle , Souris , Mutation/génétique
5.
Biol Blood Marrow Transplant ; 25(8): 1492-1497, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-30959163

RÉSUMÉ

The programmed death-1 (PD-1) axis can suppress immune surveillance against multiple myeloma (MM). We tested the safety and efficacy of pembrolizumab, an anti-PD-1 antibody, in MM after autologous hematopoietic cell transplantation (AHCT). We enrolled patients with MM who did not achieve a complete response (CR) to induction therapy. The study intervention involved a total of 9 doses of i.v. pembrolizumab, with 1 dose given every 21 days starting on day +14 post-AHCT. The primary endpoint was the rate of CR at end of treatment (EOT) in patients receiving ≥2 pembrolizumab doses. Thirty-two patients were enrolled, but 3 withdrew consent before receiving the first dose. The study was terminated early after failing to meet its interim analysis endpoint to detect a 20% difference in EOT CR rate conversion. The median patient age was 59 years. All but 1 patient received triplet induction for a median of 4 cycles (range, 2 to 7 cycles), with 69% partial response (PR) and 31% very good PR (VGPR). No grade 4/5 toxicities or graft failures occurred. Among 26 evaluable patients, 23 had an EOT evaluation, and 7 of these 23 (31%) achieved CR. Two patients had EOT serologic CR but no bone marrow confirmation (CRu), and 1 patient had no EOT evaluation. Bone marrow was minimal residual disease-negative by flow cytometry in 12 of 16 patients (75%) at day +180. With a median follow-up of 23.7 months (range, 15.1 to 33.5 months), no patient achieving EOT CR/CRu had relapsed, whereas 3 patients progressed before EOT and 1 patient progressed at 8 months after EOT VGPR. The estimated 2-year progression-free rate was 83% (95% confidence interval, 68% to 100%). Our data show that early post-AHCT pembrolizumab with lenalidomide maintenance is feasible; however, the efficacy is uncertain and requires further study. This trial was registered at ClinicalTrials.gov (NCT02331368).


Sujet(s)
Anticorps monoclonaux humanisés/administration et posologie , Transplantation de cellules souches hématopoïétiques , Déplétion lymphocytaire , Myélome multiple/thérapie , Adolescent , Adulte , Sujet âgé , Anticorps monoclonaux humanisés/effets indésirables , Autogreffes , Femelle , Études de suivi , Humains , Mâle , Adulte d'âge moyen , Myélome multiple/sang , Induction de rémission , Facteurs temps
7.
Methods Mol Biol ; 1881: 239-251, 2019.
Article de Anglais | MEDLINE | ID: mdl-30350210

RÉSUMÉ

Droplet digital PCR (ddPCR) allows precise quantitation of individual copies of DNA. Using allele-specific fluorescent oligonucleotide probes, ddPCR can enumerate both mutant and wild-type alleles enabling highly sensitive detection and quantitation of the variant allele frequency of mutated genes. In this protocol, we describe a method for using ddPCR to detect mutations in genomic DNA with a sensitivity of up to 1 in 50,000 DNA copies.


Sujet(s)
Analyse de mutations d'ADN/méthodes , Mutation , Réaction de polymérisation en chaîne/méthodes , Allèles , ADN/analyse , ADN/génétique , Génome humain , Humains , Protéines nucléaires/génétique , Nucléophosmine
8.
Blood ; 131(12): 1372-1379, 2018 03 22.
Article de Anglais | MEDLINE | ID: mdl-29437593

RÉSUMÉ

Corticosteroid resistance after acute graft-versus-host disease (SR-aGVHD) results in high morbidity and mortality after allogeneic hematopoietic cell transplantation. Current immunosuppressive therapies for SR-aGVHD provide marginal effectiveness because of poor response or excessive toxicity, primarily from infection. α1-Antitrypsin (AAT), a naturally abundant serine protease inhibitor, is capable of suppressing experimental GVHD by downmodulating inflammation and increasing ratios of regulatory (Treg) to effector T cells (Teffs). In this prospective multicenter clinical study, we sought to determine the safety and response rate of AAT administration in SR-aGVHD. Forty patients with a median age of 59 years received intravenous AAT twice weekly for 4 weeks as first-line treatment of SR-aGVHD. The primary end point was overall response rate (ORR), the proportion of patients with SR-aGVHD in complete (CR) or partial response by day 28 without addition of further immunosuppression. Treatment was well tolerated without drug-related adverse events. A significant increase in serum levels of AAT was observed after treatment. The ORR and CR rates by day 28 were 65% and 35%, respectively, and included responses in all aGVHD target organs. At day 60, responses were sustained in 73% of patients without intervening immunosuppression. Infectious mortality was 10% at 6 months and 2.5% within 30 days of last AAT infusion. Consistent with preclinical data, correlative samples showed an increase in ratio of activated Tregs to Teffs after AAT treatment. These data suggest that AAT is safe and may be potentially efficacious in treating SR-aGVHD. This trial was registered at www.clinicaltrials.gov as #NCT01700036.


Sujet(s)
Maladie du greffon contre l'hôte , alpha-1-Antitrypsine , Maladie aigüe , Administration par voie intraveineuse , Adulte , Survie sans rechute , Femelle , Maladie du greffon contre l'hôte/sang , Maladie du greffon contre l'hôte/traitement médicamenteux , Maladie du greffon contre l'hôte/mortalité , Humains , Infections/sang , Infections/traitement médicamenteux , Infections/mortalité , Mâle , Adulte d'âge moyen , Études prospectives , Taux de survie , alpha-1-Antitrypsine/administration et posologie , alpha-1-Antitrypsine/pharmacocinétique
9.
J Cell Mol Med ; 22(3): 1411-1427, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29327808

RÉSUMÉ

Acute myeloid leukaemia (AML) is a malignant disorder of the myeloid blood lineage characterized by impaired differentiation and increased proliferation of hematopoietic precursor cells. Recent technological advances have led to an improved understanding of AML biology but also uncovered the enormous cytogenetic and molecular heterogeneity of the disease. Despite this heterogeneity, AML is mostly managed by a 'one-size-fits-all' approach consisting of intensive, highly toxic induction and consolidation chemotherapy. These treatment protocols have remained largely unchanged for the past several decades and only lead to a cure in approximately 30-35% of cases. The advent of targeted therapies in chronic myeloid leukaemia and other malignancies has sparked hope to improve patient outcome in AML. However, the implementation of targeted agents in AML therapy has been unexpectedly cumbersome and remains a difficult task due to a variety of disease- and patient-specific factors. In this review, we describe current standard and investigational therapeutic strategies with a focus on targeted agents and highlight potential tools that might facilitate the development of targeted therapies for this fatal disease. The classes of agents described in this review include constitutively activated signalling pathway inhibitors, surface receptor targets, epigenetic modifiers, drugs targeting the interaction of the hematopoietic progenitor cell with the stroma and drugs that target the apoptotic machinery. The clinical context and outcome with these agents will be examined to gain insight about their optimal utilization.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques , Leucémie aigüe myéloïde/traitement médicamenteux , Médecine de précision/méthodes , Prise en charge de la maladie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Thérapie moléculaire ciblée , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes c-kit/antagonistes et inhibiteurs , Protéines proto-oncogènes c-kit/génétique , Protéines proto-oncogènes c-kit/métabolisme , Induction de rémission , Tyrosine kinase-3 de type fms/antagonistes et inhibiteurs , Tyrosine kinase-3 de type fms/génétique , Tyrosine kinase-3 de type fms/métabolisme , Protéines G ras/antagonistes et inhibiteurs , Protéines G ras/génétique , Protéines G ras/métabolisme
10.
Blood ; 130(15): 1760-1767, 2017 10 12.
Article de Anglais | MEDLINE | ID: mdl-28784598

RÉSUMÉ

The oral histone deacetylase (HDAC) inhibitor (vorinostat) is safe and results in low incidence of acute graft-versus-host disease (GVHD) after reduced-intensity conditioning, related donor hematopoietic cell transplantation (HCT). However, its safety and efficacy in preventing acute GVHD in settings of heightened clinical risk that use myeloablative conditioning, unrelated donor (URD), and methotrexate are not known. We conducted a prospective, phase 2 study in this higher-risk setting. We enrolled 37 patients to provide 80% power to detect a significant difference in grade 2 to 4 acute GVHD of 50% compared with a reduction in target to 28%. Eligibility included adults with a hematological malignancy to receive myeloablative HCT from an available 8/8-HLA matched URD. Patients received GVHD prophylaxis with tacrolimus and methotrexate. Vorinostat (100 mg twice daily) was started on day -10 and continued through day +100 post-HCT. Median age was 56 years (range, 18-69 years), and 95% had acute myelogenous leukemia or high-risk myelodysplastic syndrome. Vorinostat was safe and tolerable. The cumulative incidence of grade 2 to 4 acute GVHD at day 100 was 22%, and for grade 3 to 4 it was 8%. The cumulative incidence of chronic GVHD was 29%; relapse, nonrelapse mortality, GVHD-free relapse-free survival, and overall survival at 1 year were 19%, 16%, 47%, and 76%, respectively. Correlative analyses showed enhanced histone (H3) acetylation in peripheral blood mononuclear cells and reduced interleukin 6 (P = .028) and GVHD biomarkers (Reg3, P = .041; ST2, P = .002) at day 30 post-HCT in vorinostat-treated subjects compared with similarly treated patients who did not receive vorinostat. Vorinostat for GVHD prevention is an effective strategy that should be confirmed in a randomized phase 3 study. This trial was registered at www.clinicaltrials.gov as #NCT01790568.


Sujet(s)
Maladie du greffon contre l'hôte/traitement médicamenteux , Maladie du greffon contre l'hôte/prévention et contrôle , Transplantation de cellules souches hématopoïétiques/effets indésirables , Acides hydroxamiques/usage thérapeutique , Méthotrexate/usage thérapeutique , Tacrolimus/usage thérapeutique , Conditionnement pour greffe/effets indésirables , Donneurs non apparentés , Acétylation , Maladie aigüe , Adolescent , Adulte , Sujet âgé , Démographie , Études de faisabilité , Femelle , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Histone/métabolisme , Humains , Acides hydroxamiques/effets indésirables , Incidence , Mâle , Méthotrexate/effets indésirables , Adulte d'âge moyen , Récidive , Analyse de survie , Tacrolimus/effets indésirables , Vorinostat , Jeune adulte
11.
J Clin Invest ; 127(9): 3484-3495, 2017 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-28825596

RÉSUMÉ

Acute myelogenous leukemia (AML) frequently relapses after complete remission (CR), necessitating improved detection and phenotypic characterization of treatment-resistant residual disease. In this work, we have optimized droplet digital PCR to broadly measure mutated alleles of recurrently mutated genes in CR marrows of AML patients at levels as low as 0.002% variant allele frequency. Most gene mutations persisted in CR, albeit at highly variable and gene-dependent levels. The majority of AML cases demonstrated residual aberrant oligoclonal hematopoiesis. Importantly, we detected very rare cells (as few as 1 in 15,000) that were genomically similar to the dominant blast populations at diagnosis and were fully clonally represented at relapse, identifying these rare cells as one common source of AML relapse. Clinically, the mutant allele burden was associated with overall survival in AML, and our findings narrow the repertoire of gene mutations useful in minimal residual disease-based prognostication in AML. Overall, this work delineates rare cell populations that cause AML relapse, with direct implications for AML research directions and strategies to improve AML therapies and outcome.


Sujet(s)
Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/thérapie , Mutation , Récidive tumorale locale , Maladie résiduelle/génétique , Adulte , Sujet âgé , Allèles , Anthracyclines/administration et posologie , Moelle osseuse/anatomopathologie , Cytarabine/administration et posologie , Exome , Femelle , Hématopoïèse , Humains , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Phénotype , Pronostic , Induction de rémission , Transplantation de cellules souches , Facteurs temps , Transplantation homologue , Résultat thérapeutique , Jeune adulte
12.
Clin Cancer Res ; 22(17): 4525-35, 2016 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-27060156

RÉSUMÉ

PURPOSE: Chronic lymphocytic leukemia (CLL)-associated gene mutations that influence CLL cell fitness and chemotherapy resistance should increase in clonal representation when measured before therapy and at relapse. EXPERIMENTAL DESIGN: To uncover mutations associated with CLL relapse, we have performed whole-exome sequencing in a discovery cohort of 61 relapsed CLL patients identifying 86 recurrently mutated genes. The variant allele fractions (VAF) of 19 genes with mutations in ≥3 of 61 cases were measured in 53 paired pre- and posttreatment CLL samples sorted to purity using panel-based deep resequencing or by droplet digital PCR. RESULTS: We identify mutations in TP53 as the dominant subclonal gene driver of relapsed CLL often demonstrating substantial increases in VAFs. Subclonal mutations in SAMHD1 also recurrently demonstrated increased VAFs at relapse. Mutations in ATP10A, FAT3, FAM50A, and MGA, although infrequent, demonstrated enrichment in ≥2 cases each. In contrast, mutations in NOTCH1, SF3B1, POT1, FBXW7, MYD88, NXF1, XPO1, ZMYM3, or CHD2 were predominantly already clonal prior to therapy indicative of a pretreatment pathogenetic driver role in CLL. Quantitative analyses of clonal dynamics uncover rising, stable, and falling clones and subclones without clear evidence that gene mutations other than in TP53 and possibly SAMHD1 are frequently selected for at CLL relapse. CONCLUSIONS: Data in aggregate support a provisional categorization of CLL-associated recurrently mutated genes into three classes (i) often subclonal before therapy and strongly enriched after therapy, or, (ii) mostly clonal before therapy or without further enrichments at relapse, or, (iii) subclonal before and after therapy and enriching only in sporadic cases. Clin Cancer Res; 22(17); 4525-35. ©2016 AACR.


Sujet(s)
Évolution clonale/génétique , Leucémie chronique lymphocytaire à cellules B/génétique , Mutation , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Allèles , Marqueurs biologiques tumoraux , Évolution de la maladie , Femelle , Humains , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Mâle , Adulte d'âge moyen , Stadification tumorale , Pronostic ,
13.
Biotechniques ; 60(4): 175-6, 178, 180 passim, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-27071606

RÉSUMÉ

Digital PCR (dPCR) is gaining popularity as a DNA mutation quantification method for clinical specimens. Fragmentation prior to dPCR is required for non-fragmented genomic DNA samples; however, the effect of fragmentation on DNA analysis has not been well-studied. Here we evaluated three fragmentation methods for their effects on dPCR point mutation assay performance. Wild-type (WT) human genomic DNA was fragmented by heating, restriction digestion, or acoustic shearing using a Covaris focused-ultrasonicator. dPCR was then used to determine the limit of blank (LoB) by quantifying observed WT and mutant allele counts of the proto-oncogenes KRAS and BRAF in the WT DNA sample. DNA fragmentation by heating to 95°C, while the simplest and least expensive method, produced a high background mutation frequency for certain KRAS mutations relative to the other methods. This was due to heat-induced mutations, specifically affecting dPCR assays designed to interrogate guanine to adenine (G>A) mutations. Moreover, heat-induced fragmentation overestimated gene copy number, potentially due to denaturation and partition of single-stranded DNA into different droplets. Covaris acoustic shearing and restriction enzyme digestion showed similar LoBs and gene copy number estimates to one another. It should be noted that moderate heating, commonly used in genomic DNA extraction protocols, did not significantly increase observed KRAS mutation counts.


Sujet(s)
Analyse de mutations d'ADN/méthodes , ADN/génétique , Réaction de polymérisation en chaîne/méthodes , Fragmentation de l'ADN , Dosage génique , Chauffage , Humains , Mutation , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes p21(ras)/génétique
14.
Biol Blood Marrow Transplant ; 22(1): 54-60, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26211984

RÉSUMÉ

Despite the ongoing advent of more effective immunomodulators and proteasome inhibitors, multiple myeloma (MM) remains incurable and no effective therapy is available for advanced aggressive disease. Although allogeneic (Allo) hematopoietic cell transplantation (HCT) has a curative potential, the outcomes remain poor because of high treatment-related mortality (TRM), mostly due to regimen-related toxicities and graft-versus-host disease (GVHD) in case of myeloablative conditionings, high relapse rate in case of reduced-intensity or nonmyeloablative regimens, and possibly other unknown MM-specific issues. In an attempt to improve TRM, without compromising conditioning intensity, we prospectively explored the feasibility and efficacy of a myeloablative but reduced-toxicity conditioning regimen, consisting of fludarabine and busulfan (FluBu4; fludarabine 40 mg/m(2)/day and busulfan 3.2 mg/kg/day i.v. × 4 days) in 22 patients with high-risk or advanced refractory MM. The majority (14 of 22, 64%) had prior autologous HCT. The median HCT-specific comorbidity index score was 3 (range, 0 to 6), with 46% having a Karnofsky performance score < 80%. Ten patients had unrelated donors, 3 of whom were 7/8 HLA-loci matched. GVHD prophylaxis was tacrolimus and methotrexate in 20 (91%). Most patients had active MM at transplantation, with a partial response in 12 of 22 (46%) and stable disease in 1 of 22 (4.5%). All 22 patients tolerated the FluBu4 conditioning well, without early toxic deaths or graft failure. Common regimen-related toxicities included mild to moderate mucositis (18 of 22, 82%) and mild transient liver function abnormality (9 of 22, 41%). There were no grade 4 toxicities but grade 3 mucositis occurred in 7 of 22 patients (32%). The cumulative incidence of severe, grades III and IV acute GVHD at day 180 was 23% (95% confidence interval [CI], 10% to 47%) and that of chronic GVHD was 68% (95% CI, 46% to 88%). The cumulative incidences of TRM at 100 days, 1 year, and 3 years were 9% (95% CI, 2% to 33%), 19% (95% CI, 7% to 44%), and 29% (95% CI, 13% to 55%), respectively. Two TRMs were due to idiopathic pneumonia syndrome and 1 was due to cirrhosis. They all had decreased pre-HCT corresponding organ function, with HCT-specific comorbidity index scores of > 3. With a median follow-up of 58.7 (range, 39 to 82) months, the cumulative incidences of relapse at 1 and 3 years were 37% (95% CI, 20% to 61%) and 50% (95% CI, 29% to 75%); those for 1-year and 3-year overall survival (OS) were 58% (95% CI, 40% to 83%) and 29% (95% CI, 15% to 57%), respectively, and those for the 1-year and 3-year progression-free survivals (PFS) were 40% (95% CI, 23% to 67%) and 15% (95% CI, 5% to 42%), respectively. In summary, the use of the myeloablative FluBu4 conditioning Allo-HCT for high-risk MM resulted in decreased TRM, compared with that of Allo-HCT using conventional myeloablative regimens; however, the relapse rate was high, including in those developing moderate-to-severe chronic GVHD. This suggested a less robust graft-versus-myeloma effect against high-risk MM, thus resulting in poor PFS and OS. Nonetheless, the FluBu4 regimen may be used as a lower-TRM platform to combine with other strategies, eg, addition of an MM-targeted agent and/or maintenance therapy with these agents, to decrease relapse or progression in patients with high-risk MM.


Sujet(s)
Maladie du greffon contre l'hôte/mortalité , Maladie du greffon contre l'hôte/prévention et contrôle , Transplantation de cellules souches hématopoïétiques , Myélome multiple/mortalité , Myélome multiple/thérapie , Conditionnement pour greffe , Sujet âgé , Allogreffes , Femelle , Hôpitaux d'enseignement , Humains , Mâle , Michigan , Adulte d'âge moyen , Études prospectives , Facteurs de risque
15.
Bone Marrow Transplant ; 51(4): 511-520, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26191952

RÉSUMÉ

Allogeneic hematopoietic cell transplantation (HCT) has been increasingly used in the setting of FMS-like tyrosine kinase-3 (FLT3)-mutated AML. However, its role in conferring durable relapse-free intervals remains in question. Herein we sought to investigate FLT3 mutational status on transplant outcomes. We conducted a retrospective cohort study of 262 consecutive AML patients who underwent first-time allogeneic HCT (2008-2014), of whom 171 had undergone FLT3-ITD (internal tandem duplication) mutational testing. FLT3-mutated AML was associated with nearly twice the relapse risk (RR) compared with those without FLT3 mutation 3 years post-HCT (63% vs 37%, P<0.001) and with a shorter median time to relapse (100 vs 121 days). FLT3 mutational status remained significantly associated with this outcome after controlling for patient, disease and transplant-related risk factors (P<0.05). Multivariate analysis showed a significant association of FLT3 mutation with increased 3-year RR (hazard ratio (HR) 3.63, 95% confidence interval (CI): 2.13, 6.19, P<0.001) and inferior disease-free survival (HR 2.05, 95% CI: 1.29, 3.27, P<0.01) and overall survival (HR 1.92, 95% CI: 1.14, 3.24, P<0.05). These data demonstrate high risk of early relapse after allogeneic HCT for FLT3-mutated AML that translates into adverse disease-free and overall survival outcomes. Additional targeted and coordinated interventions are needed to maintain durable remission after allogeneic HCT in this high-risk population.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Leucémie aigüe myéloïde , Mutation , Tyrosine kinase-3 de type fms/génétique , Adolescent , Adulte , Sujet âgé , Allogreffes , Enfant , Enfant d'âge préscolaire , Survie sans rechute , Femelle , Humains , Nourrisson , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/mortalité , Leucémie aigüe myéloïde/thérapie , Mâle , Adulte d'âge moyen , Études rétrospectives , Facteurs de risque , Taux de survie
16.
Ann Hematol ; 94(6): 1033-41, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25784222

RÉSUMÉ

The optimal intensity of conditioning for allogeneic hematopoietic stem cell transplantation (HCT) in acute myeloid leukemia (AML) remains undefined. Traditionally, myeloablative conditioning regimens improve disease control, but at the risk of greater nonrelapse mortality. Because fludarabine with myeloablative doses of intravenous busulfan using pharmacokinetic monitoring has excellent tolerability, we reasoned that this regimen would limit relapse without substantially elevating toxicity when compared to reduced intensity conditioning. We retrospectively analyzed 148 consecutive AML patients in remission receiving T cell replete HCT conditioned with fludarabine and intravenous busulfan at doses defined as reduced (6.4 mg/kg; FluBu2, n = 63) or myeloablative (12.8 mg/kg; FluBu4, n = 85). Early and late nonrelapse mortality (NRM) was similar among FluBu4 and FluBu2 recipients, respectively (day + 100: 4 vs 0 %; 5 years: 19 vs 22 %; p = 0.54). NRM did not differ between FluBu4 and FluBu2 in patients >50 years of age (24 vs 22 %, p = 0.75). Relapse was lower in recipients of FluBu4 (5 years: 30 vs 49 %; p = 0.04), especially in patients with poor risk cytogenetics (22 vs 59 %; p = 0.02) and those >50 years of age (28 vs 51 %; p = 0.02). Overall survival favored FluBu4 recipients at 5 years (53 vs 34 %, p = 0.02), a finding confirmed in multivariate analysis (HR: 0.57; 95 % CI: 0.34-0.95; p = 0.03). These data suggest that myeloablative FluBu4 may provide equivalent NRM, reduced relapse, and improved survival compared to FluBu2, emphasizing the importance of busulfan dose in conditioning for AML.


Sujet(s)
Busulfan/administration et posologie , Transplantation de cellules souches hématopoïétiques , Leucémie aigüe myéloïde/thérapie , Agonistes myélo-ablatifs/administration et posologie , Conditionnement pour greffe/méthodes , Vidarabine/analogues et dérivés , Adulte , Sujet âgé , Association de médicaments , Femelle , Transplantation de cellules souches hématopoïétiques/effets indésirables , Transplantation de cellules souches hématopoïétiques/mortalité , Humains , Leucémie aigüe myéloïde/mortalité , Mâle , Adulte d'âge moyen , Induction de rémission/méthodes , Études rétrospectives , Taux de survie/tendances , Conditionnement pour greffe/mortalité , Transplantation homologue/méthodes , Transplantation homologue/mortalité , Vidarabine/administration et posologie , Jeune adulte
17.
Clin Cancer Res ; 21(9): 2045-56, 2015 May 01.
Article de Anglais | MEDLINE | ID: mdl-25652455

RÉSUMÉ

PURPOSE: Recurrent gene mutations, chromosomal translocations, and acquired genomic copy number aberrations (aCNA) have been variously associated with acute myelogenous leukemia (AML) patient outcome. However, knowledge of the co-occurrence of such lesions and the relative influence of different types of genomic alterations on clinical outcomes in AML is still evolving. EXPERIMENTAL DESIGN: We performed SNP 6.0 array-based genomic profiling of aCNA/copy neutral loss-of-heterozygosity (cnLOH) along with sequence analysis of 13 commonly mutated genes on purified leukemic blast DNA from 156 prospectively enrolled non-FAB-M3 AML patients across the clinical spectrum of de novo, secondary, and therapy-related AML. RESULTS: TP53 and RUNX1 mutations are strongly associated with the presence of SNP-A-based aCNA/cnLOH, while FLT3 and NPM1 mutations are strongly associated with the absence of aCNA/cnLOH. The presence of mutations in RUNX1, ASXL1, and TP53, elevated SNP-A-based genomic complexity, and specific recurrent aCNAs predicted failure to achieve a complete response to induction chemotherapy. The presence of ≥1 aCNA/cnLOH and higher thresholds predicted for poor long-term survival irrespective of TP53 status, and the presence of ≥1 aCNA/cnLOH added negative prognostic information to knowledge of mutations in TET2, IDH1, NPM1, DNMT3A, and RUNX1. Results of multivariate analyses support a dominant role for TP53 mutations and a role for elevated genomic complexity as predictors of short survival in AML. CONCLUSIONS: Integrated genomic profiling of a clinically relevant adult AML cohort identified genomic aberrations most associated with SNP-A-based genomic complexity, resistance to intensive induction therapies, and shortened overall survival. Identifying SNP-A-based lesions adds prognostic value to the status of several recurrently mutated genes.


Sujet(s)
Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/mortalité , Leucémie aigüe myéloïde/thérapie , Adulte , Analyse de mutations d'ADN , Femelle , Analyse de profil d'expression de gènes , Humains , Estimation de Kaplan-Meier , Perte d'hétérozygotie , Mâle , Adulte d'âge moyen , Nucléophosmine , Séquençage par oligonucléotides en batterie , Polymorphisme de nucléotide simple , Pronostic , Modèles des risques proportionnels , Transcriptome
18.
Blood ; 125(4): 668-79, 2015 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-25428220

RÉSUMÉ

Follicular lymphoma (FL) is the second most common non-Hodgkin lymphoma in the Western world. FL cell-intrinsic and cell-extrinsic factors influence FL biology and clinical outcome. To further our understanding of the genetic basis of FL, we performed whole-exome sequencing of 23 highly purified FL cases and 1 transformed FL case and expanded findings to a combined total of 114 FLs. We report recurrent mutations in the transcription factor STAT6 in 11% of FLs and identified the STAT6 amino acid residue 419 as a novel STAT6 mutation hotspot (p.419D/G, p.419D/A, and p.419D/H). FL-associated STAT6 mutations were activating, as evidenced by increased transactivation in HEK293T cell-based transfection/luciferase reporter assays, heightened interleukin-4 (IL-4) -induced activation of target genes in stable STAT6 transfected lymphoma cell lines, and elevated baseline expression levels of STAT6 target genes in primary FL B cells harboring mutant STAT6. Mechanistically, FL-associated STAT6 mutations facilitated nuclear residency of STAT6, independent of IL-4-induced STAT6-Y641 phosphorylation. Structural modeling of STAT6 based on the structure of the STAT1-DNA complex revealed that most FL-associated STAT6 mutants locate to the STAT6-DNA interface, potentially facilitating heightened interactions. The genetic and functional data combined strengthen the recognition of the IL-4/JAK/STAT6 axis as a driver of FL pathogenesis.


Sujet(s)
Noyau de la cellule/métabolisme , Régulation de l'expression des gènes tumoraux , Lymphome folliculaire/métabolisme , Mutation faux-sens , Protéines tumorales/métabolisme , Facteur de transcription STAT-6/métabolisme , Transport nucléaire actif/génétique , Lignée cellulaire tumorale , Noyau de la cellule/génétique , Étude d'association pangénomique , Cellules HEK293 , Humains , Interleukine-4/génétique , Interleukine-4/métabolisme , Janus kinases/génétique , Janus kinases/métabolisme , Lymphome folliculaire/génétique , Lymphome folliculaire/anatomopathologie , Protéines tumorales/génétique , Phosphorylation/génétique , Facteur de transcription STAT-6/génétique , Activation de la transcription/génétique
19.
Blood ; 121(2): 369-77, 2013 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-23175688

RÉSUMÉ

The frequent occurrence of persistent or relapsed disease after induction chemotherapy in AML necessitates a better understanding of the clonal relationship of AML in various disease phases. In this study, we used SNP 6.0 array-based genomic profiling of acquired copy number aberrations (aCNA) and copy neutral LOH (cnLOH) together with sequence analysis of recurrently mutated genes to characterize paired AML genomes. We analyzed 28 AML sample pairs from patients who achieved complete remission with chemotherapy and subsequently relapsed and 11 sample pairs from patients with persistent disease after induction chemotherapy. Through review of aCNA/cnLOH and gene mutation profiles in informative cases, we demonstrate that relapsed AML invariably represents re-emergence or evolution of a founder clone. Furthermore, all individual aCNA or cnLOH detected at presentation persisted at relapse indicating that this lesion type is proximally involved in AML evolution. Analysis of informative paired persistent AML disease samples uncovered cases with 2 coexisting dominant clones of which at least one was chemotherapy sensitive and one resistant, respectively. These data support the conclusion that incomplete eradication of AML founder clones rather than stochastic emergence of fully unrelated novel clones underlies AML relapse and persistence with direct implications for clinical AML research.


Sujet(s)
Évolution clonale/génétique , Leucémie aigüe myéloïde/génétique , Récidive tumorale locale/génétique , Adulte , Antinéoplasiques/usage thérapeutique , Clones cellulaires , Hybridation génomique comparative , Cytométrie en flux , Dosage génique , Humains , Hybridation fluorescente in situ , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Perte d'hétérozygotie , RT-PCR
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...