Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 52
Filtrer
1.
Leuk Lymphoma ; 65(6): 783-788, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38380861

RÉSUMÉ

STOP-CA was a multicenter, double-blind, randomized, placebo-controlled trial comparing atorvastatin to placebo in treatment-naïve lymphoma patients receiving anthracycline-based chemotherapy. We performed a preplanned subgroup to analyze the impact of atorvastatin on efficacy in patients with diffuse large B-cell lymphoma (DLBCL). Patients received rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) at standard doses for six 21-day cycles and were randomly assigned to receive atorvastatin 40 mg daily (n = 55) or placebo (n = 47) for 12 months. The complete response (CR) rate was numerically higher in the atorvastatin arm (95% [52/55] vs. 85% [40/47], p = .18), but this was not statistically significant. Adverse event rates were similar between the atorvastatin and placebo arms. In summary, atorvastatin did not result in a statistically significant improvement in the CR rate or progression-free survival, but both were numerically improved in the atorvastatin arm. These data warrant further investigation into the potential therapeutic role of atorvastatin added to anthracycline-based chemotherapies.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Atorvastatine , Cyclophosphamide , Doxorubicine , Lymphome B diffus à grandes cellules , Prednisone , Rituximab , Vincristine , Humains , Atorvastatine/usage thérapeutique , Atorvastatine/administration et posologie , Atorvastatine/effets indésirables , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/mortalité , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Prednisone/usage thérapeutique , Prednisone/effets indésirables , Prednisone/administration et posologie , Vincristine/usage thérapeutique , Vincristine/effets indésirables , Rituximab/usage thérapeutique , Rituximab/effets indésirables , Rituximab/administration et posologie , Doxorubicine/usage thérapeutique , Doxorubicine/effets indésirables , Doxorubicine/administration et posologie , Cyclophosphamide/usage thérapeutique , Cyclophosphamide/effets indésirables , Cyclophosphamide/administration et posologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Méthode en double aveugle , Résultat thérapeutique , Adulte
2.
Clin Cancer Res ; 30(11): 2514-2530, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38252421

RÉSUMÉ

PURPOSE: Develop a novel therapeutic strategy for patients with subtypes of mature T-cell and NK-cell neoplasms. EXPERIMENTAL DESIGN: Primary specimens, cell lines, patient-derived xenograft models, commercially available, and proprietary anti-KLRG1 antibodies were used for screening, target, and functional validation. RESULTS: Here we demonstrate that surface KLRG1 is highly expressed on tumor cells in subsets of patients with extranodal NK/T-cell lymphoma (ENKTCL), T-prolymphocytic leukemia (T-PLL), and gamma/delta T-cell lymphoma (G/D TCL). The majority of the CD8+/CD57+ or CD3-/CD56+ leukemic cells derived from patients with T- and NK-large granular lymphocytic leukemia (T-LGLL and NK-LGLL), respectively, expressed surface KLRG1. The humanized afucosylated anti-KLRG1 monoclonal antibody (mAb208) optimized for mouse in vivo use depleted KLRG1+ TCL cells by mechanisms of ADCC, ADCP, and CDC rather than apoptosis. mAb208 induced ADCC and ADCP of T-LGLL patient-derived CD8+/CD57+ cells ex vivo. mAb208 effected ADCC of subsets of healthy donor-derived KLRG1+ NK, CD4+, CD8+ Tem, and TemRA cells while sparing KLRG1- naïve and CD8+ Tcm cells. Treatment of cell line and TCL patient-derived xenografts with mAb208 or anti-CD47 mAb alone and in combination with the PI3K-δ/γ inhibitor duvelisib extended survival. The depletion of macrophages in vivo antagonized mAb208 efficacy. CONCLUSIONS: Our findings suggest the potential benefit of a broader treatment strategy combining therapeutic antibodies with PI3Ki for the treatment of patients with mature T-cell and NK-cell neoplasms. See related commentary by Varma and Diefenbach, p. 2300.


Sujet(s)
Lectines de type C , Récepteurs immunologiques , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Souris , Récepteurs immunologiques/antagonistes et inhibiteurs , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/immunologie , Lectines de type C/métabolisme , Lectines de type C/immunologie , Lectines de type C/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Lymphome T/immunologie , Lymphome T/anatomopathologie , Lymphome T/thérapie , Lymphome T/traitement médicamenteux , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/pharmacologie
3.
Ann Hematol ; 103(1): 185-198, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37851072

RÉSUMÉ

Antibodies targeting PD-1 or 4-1BB achieve objective responses in follicular lymphoma (FL), but only in a minority of patients. We hypothesized that targeting multiple immune receptors could overcome immune resistance and increase response rates in patients with relapsed/refractory FL. We therefore conducted a phase 1b trial testing time-limited therapy with different immunotherapy doublets targeting 4-1BB (utomilumab), OX-40 (ivuxolimab), and PD-L1 (avelumab) in combination with rituximab among patients with relapsed/refractory grade 1-3A FL. Patients were enrolled onto 2 of 3 planned cohorts (cohort 1 - rituximab/utomilumab/avelumab; cohort 2 - rituximab/ivuxolimab/utomilumab). 3+3 dose escalation was followed by dose expansion at the recommended phase 2 dose (RP2D). Twenty-four patients were enrolled (16 in cohort 1 and 9 in cohort 2, with one treated in both cohorts). No patients discontinued treatment due to adverse events and the RP2D was the highest dose level tested in both cohorts. In cohort 1, the objective and complete response rates were 44% and 19%, respectively (50% and 30%, respectively, at RP2D). In cohort 2, no responses were observed. The median progression-free survivals in cohorts 1 and 2 were 6.9 and 3.2 months, respectively. In cohort 1, higher density of PD-1+ tumor-infiltrating T-cells on baseline biopsies and lower density of 4-1BB+ and TIGIT+ T-cells in on-treatment biopsies were associated with response. Abundance of Akkermansia in stool samples was also associated with response. Our results support a possible role for 4-1BB agonist therapy in FL and suggest that features of the tumor microenvironment and stool microbiome may be associated with clinical outcomes (NCT03636503).


Sujet(s)
Antinéoplasiques , Lymphome folliculaire , Humains , Rituximab , Lymphome folliculaire/traitement médicamenteux , Récepteur-1 de mort cellulaire programmée , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Récidive tumorale locale/traitement médicamenteux , Antinéoplasiques/usage thérapeutique , Anticorps monoclonaux/effets indésirables , Immunothérapie , Microenvironnement tumoral
4.
JAMA ; 330(6): 528-536, 2023 08 08.
Article de Anglais | MEDLINE | ID: mdl-37552303

RÉSUMÉ

Importance: Anthracyclines treat a broad range of cancers. Basic and retrospective clinical data have suggested that use of atorvastatin may be associated with a reduction in cardiac dysfunction due to anthracycline use. Objective: To test whether atorvastatin is associated with a reduction in the proportion of patients with lymphoma receiving anthracyclines who develop cardiac dysfunction. Design, Setting, and Participants: Double-blind randomized clinical trial conducted at 9 academic medical centers in the US and Canada among 300 patients with lymphoma who were scheduled to receive anthracycline-based chemotherapy. Enrollment occurred between January 25, 2017, and September 10, 2021, with final follow-up on October 10, 2022. Interventions: Participants were randomized to receive atorvastatin, 40 mg/d (n = 150), or placebo (n = 150) for 12 months. Main Outcomes and Measures: The primary outcome was the proportion of participants with an absolute decline in left ventricular ejection fraction (LVEF) of ≥10% from prior to chemotherapy to a final value of <55% over 12 months. A secondary outcome was the proportion of participants with an absolute decline in LVEF of ≥5% from prior to chemotherapy to a final value of <55% over 12 months. Results: Of the 300 participants randomized (mean age, 50 [SD, 17] years; 142 women [47%]), 286 (95%) completed the trial. Among the entire cohort, the baseline mean LVEF was 63% (SD, 4.6%) and the follow-up LVEF was 58% (SD, 5.7%). Study drug adherence was noted in 91% of participants. At 12-month follow-up, 46 (15%) had a decline in LVEF of 10% or greater from prior to chemotherapy to a final value of less than 55%. The incidence of the primary end point was 9% (13/150) in the atorvastatin group and 22% (33/150) in the placebo group (P = .002). The odds of a 10% or greater decline in LVEF to a final value of less than 55% after anthracycline treatment was almost 3 times greater for participants randomized to placebo compared with those randomized to atorvastatin (odds ratio, 2.9; 95% CI, 1.4-6.4). Compared with placebo, atorvastatin also reduced the incidence of the secondary end point (13% vs 29%; P = .001). There were 13 adjudicated heart failure events (4%) over 24 months of follow-up. There was no difference in the rates of incident heart failure between study groups (3% with atorvastatin, 6% with placebo; P = .26). The number of serious related adverse events was low and similar between groups. Conclusions and Relevance: Among patients with lymphoma treated with anthracycline-based chemotherapy, atorvastatin reduced the incidence of cardiac dysfunction. This finding may support the use of atorvastatin in patients with lymphoma at high risk of cardiac dysfunction due to anthracycline use. Trial Registration: ClinicalTrials.gov Identifier: NCT02943590.


Sujet(s)
Anthracyclines , Antibiotiques antinéoplasiques , Atorvastatine , Agents cardiovasculaires , Cardiopathies , Lymphomes , Femelle , Humains , Adulte d'âge moyen , Anthracyclines/effets indésirables , Anthracyclines/usage thérapeutique , Antibiotiques antinéoplasiques/effets indésirables , Antibiotiques antinéoplasiques/usage thérapeutique , Atorvastatine/usage thérapeutique , Méthode en double aveugle , Défaillance cardiaque/étiologie , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/prévention et contrôle , Études rétrospectives , Débit systolique , Fonction ventriculaire gauche , Agents cardiovasculaires/usage thérapeutique , Lymphomes/traitement médicamenteux , Cardiopathies/induit chimiquement , Cardiopathies/physiopathologie , Cardiopathies/prévention et contrôle , Études de suivi , Mâle , Adulte , Sujet âgé
5.
Blood ; 142(7): 621-628, 2023 08 17.
Article de Anglais | MEDLINE | ID: mdl-37319432

RÉSUMÉ

Autologous stem cell transplantation (ASCT) is often used as consolidation for several subtypes of peripheral T-cell lymphoma (PTCL) in first remission. However, many patients relapse after ASCT and have a very poor prognosis. There are no approved treatment options for posttransplantation maintenance or consolidation in PTCL. PD-1 blockade has demonstrated some efficacy for patients with PTCL. We, therefore, conducted a phase 2 multicenter study of the anti-PD-1 monoclonal antibody pembrolizumab after ASCT in patients with PTCL in first remission. Pembrolizumab was administered at 200 mg IV every 3 weeks for up to 8 cycles within 21 days from post-ASCT discharge (and within 60 days of stem cell infusion). The primary end point was progression-free survival (PFS) at 18 months after ASCT. Twenty-one patients were treated in this study and 67% (n = 14) completed 8 cycles of treatment. Among all patients who were evaluable, 13 of 21 were alive and achieved PFS at 18 months after ASCT, meeting the study's primary end point. The estimated 18-month PFS was 83.6% (95% confidence interval [CI], 68-100), and overall survival 94.4% (95% CI, 84-100). The toxicity profile was consistent with the known toxicity profile of pembrolizumab, with no grade 5 toxicities. In conclusion, PD-1 blockade after ASCT with pembrolizumab is feasible with a favorable safety profile and promising activity, supporting further confirmatory studies. This trial was registered at www.clinicaltrials.gov as #NCT02362997.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Lymphome T périphérique , Humains , Survie sans rechute , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Récidive tumorale locale/étiologie , Transplantation autologue , Lymphome T périphérique/traitement médicamenteux , Lymphocytes T/anatomopathologie , Transplantation de cellules souches
6.
Leukemia ; 37(4): 835-842, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36717653

RÉSUMÉ

This study investigated ibrutinib plus obinutuzumab in relapsed/refractory CLL, evaluating tolerability of 3 sequencing regimens as well as overall safety and efficacy. Fifty-two patients were initially randomized 1:1:1 to receive either obinutuzumab 1 month before ibrutinib initiation, ibrutinib 1 month prior to obinutuzumab initiation, or to start both drugs concomitantly. Higher rates of infusion-related reactions were observed with the first sequence, and only the latter 2 cohorts were expanded. Grade 4 hematologic toxicity was uncommon, and notable all-grade non-hematologic toxicities included bruising (58%), hypertension (46%), arthralgia (38%), diarrhea (37%), transaminitis (35%), atrial fibrillation (21%), and serious infection (17%). Best overall response rate was 96% (including 40% CR and 56% PR). Best rates of undetectable minimal residual disease in peripheral blood and bone marrow were 27% and 19%, respectively. With a median follow-up of 41.5 months, four-year progression-free and overall survival rates are 74% and 93%, respectively. Correlative studies demonstrated that serum CCL4 and CXCL13 levels were associated with clinical response, and BH3 profiling revealed increased BCL-2 and BCL-xL dependence in CLL cells from patients on treatment. Overall, ibrutinib plus obinutuzumab was highly active, with a manageable safety profile, supporting further investigation of this type of approach in relapsed/refractory CLL.


Sujet(s)
Leucémie chronique lymphocytaire à cellules B , Humains , Anticorps monoclonaux humanisés/effets indésirables , Pipéridines/usage thérapeutique , Récidive , Protocoles de polychimiothérapie antinéoplasique/effets indésirables
10.
Ann Hematol ; 100(10): 2529-2539, 2021 Oct.
Article de Anglais | MEDLINE | ID: mdl-34304287

RÉSUMÉ

We conducted a phase II clinical trial to develop an autologous EBV-specific T cell product (baltaleucel T) for advanced, relapsed ENKTL. Among 47 patients who provided whole blood starting material for manufacturing the product, 15 patients received a median of 4 doses of baltaleucel T. Thirty-two (68%) patients did not receive baltaleucel-T due to manufacturing failure, rapid disease progression, and death. Of the 15 patients, 10 patients had measurable disease at baseline (salvage cohort), and 5 patients had no disease at baseline assessment (adjuvant cohort). In the 15 patients, the median follow-up duration was 10.2 months (range 2.0-23.5 months), median progression-free survival (PFS) was 3.9 months, and the median overall survival (OS) was not reached. Patients in the salvage cohort achieved a 30% complete response (CR) and a 50% overall response rate (ORR). In the adjuvant cohort, disease progression was reported in three patients and two patients did not relapse during study follow-up. When we compared survival outcomes of seven responders and eight non-responders, the PFS (P = 0.001) and OS (P = 0.014) of responders proved statistically superior to that of non-responders. Baltaleucel-T was well tolerated. We have performed a phase II clinical trial of autologous EBV-specific T cell treatment (baltaleucel-T) in R/R ENKTL. Autologous EBV-specific T cells were well tolerated and demonstrated single-agent activity in R/R ENTKL.


Sujet(s)
Infections à virus Epstein-Barr/immunologie , Herpèsvirus humain de type 4/immunologie , Lymphome T-NK extraganglionnaire/immunologie , Lymphome T-NK extraganglionnaire/thérapie , Lymphocytes T/immunologie , Adulte , Sujet âgé , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/thérapie , Femelle , Humains , Immunothérapie adoptive , Lymphome T-NK extraganglionnaire/complications , Mâle , Adulte d'âge moyen , Lymphocytes T/transplantation , Résultat thérapeutique , Jeune adulte
11.
Am J Hematol ; 96(4): E95-E98, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33373063
12.
Blood ; 137(10): 1353-1364, 2021 03 11.
Article de Anglais | MEDLINE | ID: mdl-32871584

RÉSUMÉ

T-cell/histiocyte-rich large B-cell lymphoma (TCRLBCL) is an aggressive variant of diffuse large B-cell lymphoma (DLBCL) characterized by rare malignant B cells within a robust but ineffective immune cell infiltrate. The mechanistic basis of immune escape in TCRLBCL is poorly defined and not targeted therapeutically. We performed a genetic and quantitative spatial analysis of the PD-1/PD-L1 pathway in a multi-institutional cohort of TCRLBCLs and found that malignant B cells harbored PD-L1/PD-L2 copy gain or amplification in 64% of cases, which was associated with increased PD-L1 expression (P = .0111). By directed and unsupervised spatial analyses of multiparametric cell phenotypic data within the tumor microenvironment, we found that TCRLBCL is characterized by tumor-immune "neighborhoods" in which malignant B cells are surrounded by exceptionally high numbers of PD-L1-expressing TAMs and PD-1+ T cells. Furthermore, unbiased clustering of spatially resolved immune signatures distinguished TCRLBCL from related subtypes of B-cell lymphoma, including classic Hodgkin lymphoma (cHL) and DLBCL-NOS. Finally, we observed clinical responses to PD-1 blockade in 3 of 5 patients with relapsed/refractory TCRLBCL who were enrolled in clinical trials for refractory hematologic malignancies (NCT03316573; NCT01953692), including 2 complete responses and 1 partial response. Taken together, these data implicate PD-1 signaling as an immune escape pathway in TCRLBCL and also support the potential utility of spatially resolved immune signatures to aid the diagnostic classification and immunotherapeutic prioritization of diverse tumor types.


Sujet(s)
Histiocytes/immunologie , Lymphome B diffus à grandes cellules/immunologie , Récepteur-1 de mort cellulaire programmée/immunologie , Lymphocytes T/immunologie , Échappement de la tumeur à la surveillance immunitaire , Antigène CD274/analyse , Antigène CD274/immunologie , Histiocytes/anatomopathologie , Humains , Lymphome B diffus à grandes cellules/anatomopathologie , Récepteur-1 de mort cellulaire programmée/analyse , Lymphocytes T/anatomopathologie
13.
J Drugs Dermatol ; 19(8): 803-805, 2020 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-32845598

RÉSUMÉ

Langerhans cell histiocytosis (LCH) limited to the skin is rare in adult patients. Given the challenges of prospective clinical trials for this rare disease, there is paucity in data to guide the management of cutaneous LCH. Topical nitrogen mustard is a possible treatment for cutaneous LCH with positive responses in five known adult cases in the literature. In this report, we present two adult patients with recalcitrant cutaneous LCH and no evidence of systemic involvement who had rapid and complete response on topical nitrogen mustard therapy. We provide support for topical nitrogen mustard as a treatment option for primary cutaneous LCH which may spare patients from requiring systemic immunosuppressive treatments. J Drugs Dermatol. 2020;19(8):803-805. doi:10.36849/JDD.2020.4943.


Sujet(s)
Agents alcoylants/administration et posologie , Histiocytose à cellules de Langerhans/traitement médicamenteux , Chlorméthine/administration et posologie , Maladies de la peau/traitement médicamenteux , Administration par voie cutanée , Sujet âgé , Sujet âgé de 80 ans ou plus , Biopsie , Femelle , Histiocytose à cellules de Langerhans/diagnostic , Histiocytose à cellules de Langerhans/anatomopathologie , Humains , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Maladies de la peau/diagnostic , Maladies de la peau/anatomopathologie , Résultat thérapeutique
14.
Blood ; 135(22): 1929-1945, 2020 05 28.
Article de Anglais | MEDLINE | ID: mdl-32187362

RÉSUMÉ

Erdheim-Chester disease (ECD) is a rare histiocytosis that was recently recognized as a neoplastic disorder owing to the discovery of recurrent activating MAPK (RAS-RAF-MEK-ERK) pathway mutations. Typical findings of ECD include central diabetes insipidus, restrictive pericarditis, perinephric fibrosis, and sclerotic bone lesions. The histopathologic diagnosis of ECD is often challenging due to nonspecific inflammatory and fibrotic findings on histopathologic review of tissue specimens. Additionally, the association of ECD with unusual tissue tropism and an insidious onset often results in diagnostic errors and delays. Most patients with ECD require treatment, except for a minority of patients with minimally symptomatic single-organ disease. The first ECD consensus guidelines were published in 2014 on behalf of the physicians and researchers within the Erdheim-Chester Disease Global Alliance. With the recent molecular discoveries and the approval of the first targeted therapy (vemurafenib) for BRAF-V600-mutant ECD, there is a need for updated clinical practice guidelines to optimize the diagnosis and treatment of this disease. This document presents consensus recommendations that resulted from the International Medical Symposia on ECD in 2017 and 2019. Herein, we include the guidelines for the clinical, laboratory, histologic, and radiographic evaluation of ECD patients along with treatment recommendations based on our clinical experience and review of literature in the molecular era.


Sujet(s)
Maladie d'Erdheim-Chester/diagnostic , Maladie d'Erdheim-Chester/thérapie , Essais cliniques comme sujet , Maladie d'Erdheim-Chester/génétique , Femelle , Histiocytose à cellules de Langerhans/diagnostic , Histiocytose à cellules de Langerhans/génétique , Histiocytose à cellules de Langerhans/thérapie , Humains , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/génétique , Mâle , Thérapie moléculaire ciblée , Mutation , Pronostic , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/génétique
15.
Blood Adv ; 4(1): 122-126, 2020 01 14.
Article de Anglais | MEDLINE | ID: mdl-31917843

RÉSUMÉ

Disease relapse remains the leading cause of failure after autologous stem cell transplantation (ASCT) for patients with relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL). We conducted a phase 2, multicenter, single-arm study of the anti-PD-1 monoclonal antibody pembrolizumab given after ASCT in patients with chemosensitive DLBCL, hypothesizing that it would improve the progression-free survival (PFS) at 18 months after ASCT (primary endpoint) from 60% to 80%. Pembrolizumab was administered at 200 mg IV every 3 weeks for up to 8 cycles, starting within 21 days of post-ASCT discharge. Twenty-nine patients were treated on this study; 62% completed all 8 cycles. Seventy-nine percent of patients experienced at least one grade 3 or higher adverse event, and 34% experienced at least one grade 2 or higher immune-related adverse event. Overall, 59% of patients were alive and progression free at 18 months, which did not meet the primary endpoint. The 18-month overall survival was 93%. In conclusion, pembrolizumab was successfully administered as post-ASCT consolidation in patients with R/R DLBCL, but the PFS did not meet the protocol-specific primary objective and therefore does not support a larger confirmatory study. This trial was registered at www.clinicaltrials.gov as #NCT02362997.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Lymphome B diffus à grandes cellules , Humains , Lymphome B diffus à grandes cellules/traitement médicamenteux , Récepteur-1 de mort cellulaire programmée , Transplantation autologue
16.
Blood ; 134(14): 1132-1143, 2019 10 03.
Article de Anglais | MEDLINE | ID: mdl-31292114

RÉSUMÉ

T-cell prolymphocytic leukemia (T-PLL) is a rare, mature T-cell neoplasm with a heterogeneous clinical course. With the advent of novel treatment options that will potentially change the management of patients with T-PLL, it has become necessary to produce consensus guidelines for the design and conduct of clinical trials. The T-PLL International Study group (TPLL-ISG) set out to define standardized criteria for diagnosis, treatment indication, and evaluation of response. These criteria will facilitate comparison of results from clinical trials in T-PLL, and will thus support clinical decision making, as well as the approval of new therapeutics by healthcare authorities.


Sujet(s)
Leucémie prolymphocytaire à cellules T/diagnostic , Leucémie prolymphocytaire à cellules T/thérapie , Moelle osseuse/anatomopathologie , Prise en charge de la maladie , Régulation de l'expression des gènes dans la leucémie , Humains , Immunophénotypage , Leucémie prolymphocytaire à cellules T/génétique , Leucémie prolymphocytaire à cellules T/anatomopathologie , Mutation , Stadification tumorale , Lymphocytes T/anatomopathologie
17.
Hematol Oncol Clin North Am ; 33(4): 657-668, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31229161

RÉSUMÉ

Therapeutic advances for peripheral T-cell non-Hodgkin lymphoma (PTCL) have lagged behind their B-cell NHL counterparts in part because novel agents to treat PTCL have been developed empirically. The recent clinical success of brentuximab-vedotin suggests that novel therapies for PTCL can significantly improve outcomes when properly targeted. Aberrancies in T-cell receptor, Jak/STAT, and DNA methylation pathways play critical roles in T-NHL pathogenesis based on genomic studies and preclinical experimental validation. New strategies targeting these pathways in patients with PTCL are underway, and this clinical trial experience will possibly contribute to additional improvements in outcome for patients with these diseases.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Brentuximab védotine/usage thérapeutique , Immunoconjugués/usage thérapeutique , Lymphome T périphérique/thérapie , Essais cliniques comme sujet , Humains , Immunothérapie , Lymphome T périphérique/génétique , Lymphome T périphérique/anatomopathologie , Thérapie moléculaire ciblée , Transplantation de cellules souches
18.
Blood ; 134(1): 22-29, 2019 07 04.
Article de Anglais | MEDLINE | ID: mdl-30952672

RÉSUMÉ

Autologous stem cell transplantation (ASCT) remains the standard of care for patients with relapsed/refractory (RR) classical Hodgkin lymphoma (cHL) who respond to salvage chemotherapy. However, relapse after ASCT remains a frequent cause of treatment failure, with poor subsequent prognosis. Because cHL is uniquely vulnerable to programmed cell death-1 (PD-1) blockade, PD-1 blockade given as consolidation after ASCT could improve ASCT outcomes. We therefore conducted a multicohort phase 2 study of pembrolizumab in patients with RR cHL after ASCT, hypothesizing that it would improve the progression-free survival (PFS) at 18 months after ASCT (primary end point) from 60% to 80%. Pembrolizumab was administered at 200 mg IV every 3 weeks for up to 8 cycles, starting within 21 days of post-ASCT discharge. Thirty patients were treated on this study. The median age was 33 years, and 90% were high-risk by clinical criteria. Seventy-seven percent completed all 8 cycles. Toxicity was manageable, with 30% of patients experiencing at least 1 grade 3 or higher adverse event (AE), and 40% at least 1 grade 2 or higher immune-related AE. Two patients were lost to follow-up in complete remission at 12 months. The PFS at 18 months for the 28 evaluable patients was 82%, meeting the primary end point. The 18-month overall survival was 100%. In conclusion, pembrolizumab was successfully administered as post-ASCT consolidation in patients with RR cHL, and resulted in a promising PFS in a high-risk patient cohort, supporting the testing of this strategy in a randomized trial. This trial was registered at www.clinicaltrials.gov as #NCT02362997.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques immunologiques/usage thérapeutique , Maladie de Hodgkin/traitement médicamenteux , Adulte , Sujet âgé , Chimiothérapie de consolidation/méthodes , Survie sans rechute , Femelle , Transplantation de cellules souches hématopoïétiques , Maladie de Hodgkin/mortalité , Maladie de Hodgkin/chirurgie , Humains , Mâle , Adulte d'âge moyen , Récepteur-1 de mort cellulaire programmée/immunologie , Thérapie de rattrapage/méthodes , Transplantation autologue
19.
J Clin Oncol ; 37(11): 912-922, 2019 04 10.
Article de Anglais | MEDLINE | ID: mdl-30742566

RÉSUMÉ

PURPOSE: Indolent non-Hodgkin lymphoma (iNHL) remains largely incurable and often requires multiple lines of treatment after becoming refractory to standard therapies. Duvelisib was approved by the Food and Drug Administration for relapsed or refractory (RR) chronic lymphocytic leukemia or small lymphocytic lymphoma (SLL) and RR follicular lymphoma (FL) after two or more prior systemic therapies. On the basis of the activity of duvelisib, a first-in-class oral dual inhibitor of phosphoinositide 3-kinase-δ,-γ, in RR iNHL in a phase I study, the safety and efficacy of duvelisib monotherapy was evaluated in iNHL refractory to rituximab and either chemotherapy or radioimmunotherapy. PATIENTS AND METHODS: Eligible patients had measurable iNHL (FL, SLL, or marginal zone B-cell lymphoma) double refractory to rituximab (monotherapy or in combination) and to either chemotherapy or radioimmunotherapy. All were treated with duvelisib 25 mg orally twice daily in 28-day cycles until progression, unacceptable toxicity, or death. The primary end point was overall response rate (ORR) using the revised International Working Group criteria for malignant lymphoma. RESULTS: This open-label, global phase II trial enrolled 129 patients (median age, 65 years; median of three prior lines of therapy) with an ORR of 47.3% (SLL, 67.9%; FL, 42.2%; MZL, 38.9%). The estimated median duration of response was 10 months, and the estimated median progression-free survival was 9.5 months. The most frequent any-grade treatment-emergent adverse events (TEAEs) were diarrhea (48.8%), nausea (29.5%), neutropenia (28.7%), fatigue (27.9%), and cough (27.1%). Among the 88.4% of patients with at least one grade 3 or greater TEAE, the most common TEAEs were neutropenia (24.8%), diarrhea (14.7%), anemia (14.7%), and thrombocytopenia (11.6%). CONCLUSION: In the DYNAMO study, oral duvelisib monotherapy demonstrated clinically meaningful activity and a manageable safety profile in heavily pretreated, double-refractory iNHL, consistent with previous observations. Duvelisib may provide a new oral treatment option for this patient population of which many are elderly and in need of additional therapies.


Sujet(s)
Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Antienzymes/usage thérapeutique , Isoquinoléines/usage thérapeutique , Lymphome malin non hodgkinien/traitement médicamenteux , Purines/usage thérapeutique , Administration par voie orale , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Anti-inflammatoires non stéroïdiens/effets indésirables , Anti-inflammatoires non stéroïdiens/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Diarrhée/induit chimiquement , Calendrier d'administration des médicaments , Antienzymes/administration et posologie , Antienzymes/effets indésirables , Femelle , Humains , Isoquinoléines/administration et posologie , Isoquinoléines/effets indésirables , Estimation de Kaplan-Meier , Lymphome malin non hodgkinien/anatomopathologie , Mâle , Adulte d'âge moyen , Récidive tumorale locale , Phosphatidylinositol 3-kinases/métabolisme , Purines/administration et posologie , Purines/effets indésirables , Rituximab/administration et posologie
20.
Aesthet Surg J ; 39(Suppl_1): S3-S13, 2019 01 31.
Article de Anglais | MEDLINE | ID: mdl-30715173

RÉSUMÉ

National Comprehensive Cancer Network (NCCN) guidelines represent the consensus standard of care for diagnosis and management of the majority of known cancers. NCCN guidelines on breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) have been recognized by the US Food and Drug Administration and widely advocated by national specialty societies. Consensus guidelines have helped create a treatment standardization for BIA-ALCL at all stages of disease. NCCN guidelines are evidence-based where possible and utilize expert consensus opinion to fill in gaps that may exist. NCCN undergoes annual panel review by multidisciplinary faculty members, and this article represents the most up-to-date 2019 guidelines. Recommendations focus on parameters for achieving reliable diagnosis and disease management and emphasize the critical role for complete surgical ablation. Suggestions for adjunct treatments and chemotherapy regimens are included for advanced BIA-ALCL with lymph node involvement. BIA-ALCL recurrence and management of unresectable disease, and organ metastasis are addressed. Adherence to recognized BIA-ALCL guidelines ensures patients receive the most current efficacious treatment available.


Sujet(s)
Implants mammaires/effets indésirables , Lymphome à grandes cellules anaplasiques/étiologie , Femelle , Humains , Métastase lymphatique , Lymphome à grandes cellules anaplasiques/diagnostic , Lymphome à grandes cellules anaplasiques/thérapie , Récidive tumorale locale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...