Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtrer
1.
Sci Rep ; 11(1): 2306, 2021 01 27.
Article de Anglais | MEDLINE | ID: mdl-33504881

RÉSUMÉ

Evofosfamide (Evo or TH302) is a hypoxia-activated prodrug which is reduced leading to the release of alkylating agent bromo-isophosphoramide mustard, which has shown safety and signals of efficacy in a prior phase 1 study in recurrent glioblastoma. We performed a dual center single-arm Phase II study to expand on the safety and efficacy of Evo plus bevacizumab in bevacizumab refractory glioblastoma. 33 patients with bevacizumab refractory GBM received Evo 670 mg/m2 in combination with Bevacizumab 10 mg/kg IV every 2 weeks. Assessments included adverse events, response, and survival. Median age of patients was 47 (range 19-76) and 24 (69%) were male. At the time of study entry, 9 (26%) had ongoing corticosteroid use. ECOG performance status was 0 or 1 in 83% of patients. Patients were mostly heavily pretreated with 77% have three or more prior regimens. A total of 12 patients (36%) suffered grade 3-4 drug associated adverse event (AE); no grade 5 AE were reported. Of the 33 evaluable patients, best response was PR in 3 (9%), SD in 14 (43%), and PD in 16 (48%) with responses confirmed by a second reviewer. Median time to progression of disease was 53 days (95% CI 42-113) and Median time to death was 129 days (95% CI 86-199 days). Progression free survival at 4 months (PFS-4) on Evo-Bev was 31%, which was a statistically significant improvement over the historical rate of 3%. The median overall survival of patients receiving Evo-Bevacizumab was 4.6 months (95% CI 2.9-6.6). The progression free survival of patients on Evo-Bevacizumab met the primary endpoint of progression free survival at 4 months of 31%, although the clinical significance of this may be limited. Given the patient population and Phase II design, these clinical outcomes will need further validation.


Sujet(s)
Bévacizumab/usage thérapeutique , Glioblastome/traitement médicamenteux , Nitroimidazoles/usage thérapeutique , Moutardes phosphoramide/usage thérapeutique , Adulte , Sujet âgé , Femelle , Glioblastome/mortalité , Glioblastome/anatomopathologie , Humains , Mâle , Adulte d'âge moyen , Jeune adulte
2.
Neuro Oncol ; 20(9): 1231-1239, 2018 08 02.
Article de Anglais | MEDLINE | ID: mdl-29415215

RÉSUMÉ

Background: Anti-angiogenic therapy is known to induce a greater degree of hypoxia, including in glioblastoma (GBM). Evofosfamide (Evo) is a hypoxia-activated prodrug which is reduced, leading to the release of the alkylating agent bromo-isophosphoramide mustard. We assessed the safety, tolerability, preliminary efficacy, and biomarkers of Evo plus bevacizumab (Bev) in Bev-refractory GBM. Methods: Twenty-eight patients with Bev-refractory GBM were enrolled in a dose escalation study receiving from 240 mg/m2 (cohort 1) to 670 mg/m2 (cohort 4) of Evo every 2 weeks in combination with Bev. Patients deemed surgical candidates underwent a single dose of Evo or placebo with pimonidazole immediately prior to surgery for biomarker evaluation, followed by dose escalation upon recovery. Assessments included adverse events, response, and survival. Results: Evo plus Bev was well tolerated up to and including the maximum dose of 670 mg/m2, which was determined to be the recommended phase II dose. Overall response rate was 17.4%, with disease control (complete response, partial response, and stable disease) observed in 14 (60.9%) of the 23 patients. The ratio of enhancement to non-enhancement was significant on log-rank analysis with time to progression (P = 0.023), with patients having a ratio of less than 0.37 showing a median progression-free survival of 98 days versus 56 days for those with more enhancement. Conclusions: Evo plus Bev was well tolerated in patients with Bev-refractory GBM, with preliminary evidence of activity that merits further investigation.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Résistance aux médicaments antinéoplasiques , Glioblastome/traitement médicamenteux , Hypoxie , Récidive tumorale locale/traitement médicamenteux , Thérapie de rattrapage , Adulte , Sujet âgé , Bévacizumab/administration et posologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/chirurgie , Association thérapeutique , Femelle , Études de suivi , Glioblastome/anatomopathologie , Glioblastome/chirurgie , Humains , Mâle , Adulte d'âge moyen , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/chirurgie , Nitroimidazoles/administration et posologie , Moutardes phosphoramide/administration et posologie , Pronostic
3.
Neuro Oncol ; 20(6): 838-847, 2018 05 18.
Article de Anglais | MEDLINE | ID: mdl-29077941

RÉSUMÉ

Background: We recently reported an acceptable safety and pharmacokinetic profile of depatuxizumab mafodotin (depatux-m), formerly called ABT-414, plus radiation and temozolomide in newly diagnosed glioblastoma (arm A). The purpose of this study was to evaluate the safety and pharmacokinetics of depatux-m, either in combination with temozolomide in newly diagnosed or recurrent glioblastoma (arm B) or as monotherapy in recurrent glioblastoma (arm C). Methods: In this multicenter phase I dose escalation study, patients received depatux-m (0.5-1.5 mg/kg in arm B, 1.25 mg/kg in arm C) every 2 weeks by intravenous infusion. Maximum tolerated dose (MTD), recommended phase II dose (RP2D), and preliminary efficacy were also determined. Results: Thirty-eight patients were enrolled as of March 1, 2016. The most frequent toxicities were ocular, occurring in 35/38 (92%) patients. Keratitis was the most common grade 3 adverse event observed in 6/38 (16%) patients; thrombocytopenia was the most common grade 4 event seen in 5/38 (13%) patients. The MTD was set at 1.5 mg/kg in arm B and was not reached in arm C. RP2D was declared as 1.25 mg/kg for both arms. Depatux-m demonstrated a linear pharmacokinetic profile. In recurrent glioblastoma patients, the progression-free survival (PFS) rate at 6 months was 30.8% and the median overall survival was 10.7 months. Best Response Assessment in Neuro-Oncology responses were 1 complete and 2 partial responses. Conclusion: Depatux-m alone or in combination with temozolomide demonstrated an acceptable safety and pharmacokinetic profile in glioblastoma. Further studies are currently under way to evaluate its efficacy in newly diagnosed (NCT02573324) and recurrent glioblastoma (NCT02343406).


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Glioblastome/traitement médicamenteux , Adulte , Sujet âgé , Anticorps monoclonaux humanisés/administration et posologie , Tumeurs du cerveau/anatomopathologie , Femelle , Études de suivi , Glioblastome/anatomopathologie , Humains , Immunoconjugués/administration et posologie , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Pronostic , Sécurité , Taux de survie , Témozolomide/administration et posologie , Distribution tissulaire , Jeune adulte
4.
Neuro Oncol ; 19(7): 965-975, 2017 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-28039367

RÉSUMÉ

BACKGROUND: The purpose of this study was to determine the maximum tolerated dose (MTD), recommended phase II dose (RPTD), safety, and pharmacokinetics of ABT-414 plus radiation and temozolomide in newly diagnosed glioblastoma. ABT-414 is a first-in-class, tumor-specific antibody-drug conjugate that preferentially targets tumors expressing overactive epidermal growth factor receptor (EGFR). METHODS: In this multicenter phase I study, patients received 0.5-3.2 mg/kg ABT-414 every 2 weeks by intravenous infusion. EGFR alterations, O6-methylguanine-DNA methyltransferase (MGMT) promoter hypermethylation, and isocitrate dehydrogenase (IDH1) gene mutations were assessed in patient tumors. Distinct prognostic classes were assigned to patients based on a Molecular Classification Predictor model. RESULTS: As of January 7, 2016, forty-five patients were enrolled to receive ABT-414 plus radiation and temozolomide. The most common treatment emergent adverse events were ocular: blurred vision, dry eye, keratitis, photophobia, and eye pain. Ocular toxicity at any grade occurred in 40 patients and at grades 3/4 in 12 patients. RPTD and MTD were set at 2 mg/kg and 2.4 mg/kg, respectively. Among 38 patients with pretreatment tumor tested centrally, 39% harbored EGFR amplification, of which 73% had EGFRvIII mutation. Among patients with available tumor tissue (n = 30), 30% showed MGMT promoter methylation and none had IDH1 mutations. ABT-414 demonstrated an approximately dose proportional pharmacokinetic profile. The median duration of progression-free survival was 6.1 months; median overall survival has not been reached. CONCLUSION: ABT-414 plus chemoradiation demonstrated an acceptable safety and pharmacokinetic profile in newly diagnosed glioblastoma. Randomized studies are ongoing to determine efficacy in newly diagnosed (NCT02573324) and recurrent glioblastoma (NCT02343406).


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/radiothérapie , Dacarbazine/analogues et dérivés , Glioblastome/traitement médicamenteux , Glioblastome/radiothérapie , Immunoconjugués/usage thérapeutique , Adulte , Sujet âgé , Anticorps monoclonaux humanisés/pharmacologie , Antinéoplasiques/pharmacologie , Marqueurs biologiques tumoraux/métabolisme , Dacarbazine/pharmacologie , Dacarbazine/usage thérapeutique , Survie sans rechute , Association de médicaments , Femelle , Humains , Immunoconjugués/pharmacologie , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Témozolomide , Résultat thérapeutique
5.
Invest New Drugs ; 33(1): 247-53, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25388940

RÉSUMÉ

VEGF signaling through VEGFR-2 is the major factor in glioblastoma angiogenesis. CT-322, a pegylated protein engineered from the 10th type III human fibronectin domain, binds the VEGFR-2 extracellular domain with high specificity and affinity to block VEGF-induced VEGFR-2 signaling. This study evaluated CT-322 in an open-label run-in/phase 2 setting to assess its efficacy and safety in recurrent glioblastoma. Eligible patients had 1st, 2nd or 3rd recurrence of glioblastoma with measurable tumor on MRI and no prior anti-angiogenic therapy. The initial CT-322 dose was 1 mg/kg IV weekly, with plans to escalate subsequent patients to 2 mg/kg weekly if tolerated; within each CT-322 dose cohort, patients were randomized to ±irinotecan IV semiweekly. The primary endpoint was 6-month progression-free survival (PFS-6). Sixty-three patients with a median age of 56 were treated, the majority at first recurrence. One-third experienced serious adverse events, of which four were at least possibly related to study treatment (two intracranial hemorrhages and two infusion reactions). Twenty-nine percent of subjects developed treatment-emergent hypertension. The PFS-6 rate in the CT-322 monotherapy groups was 18.6 and 0.0 % in the 1 and 2 mg/kg treatment groups, respectively; results from the 2 mg/kg group indicated that the null hypothesis that PFS-6 ≤12 % could not be rejected. The study was terminated prior to reaching the planned enrollment for all treatment groups because data from the completed CT-322 2 mg/kg monotherapy treatment arm revealed insufficient efficacy. Despite biological activity and a tolerable side effect profile, CT-322 failed to meet the prespecified threshold for efficacy in recurrent glioblastoma.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Fibronectines/usage thérapeutique , Glioblastome/traitement médicamenteux , Fragments peptidiques/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Adulte , Sujet âgé , Antinéoplasiques/effets indésirables , Tumeurs du cerveau/génétique , Survie sans rechute , Femelle , Fibronectines/effets indésirables , Glioblastome/génétique , Glucuronosyltransferase/génétique , Humains , Mâle , Adulte d'âge moyen , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/génétique , Fragments peptidiques/effets indésirables , Polymorphisme génétique , Inhibiteurs de protéines kinases/effets indésirables
6.
Transplantation ; 75(1): 118-26, 2003 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-12544883

RÉSUMÉ

BACKGROUND: Graft-versus-host disease (GVHD) is an important, underdiagnosed cause of mortality associated with liver transplantation. We identified 12 cases of GVHD among 1,082 liver transplantations performed in patients at our institution between 1991 and 1998. Patients typically developed fever, skin rash, diarrhea, or pancytopenia within 2 to 6 weeks after their transplant. Treatment generally involved increased immune suppression and hematopoietic cytokines (granulocyte colony stimulating factor, granulocyte monocyte colony stimulating factor); however, all but one patient died, most often from sepsis. Early in its course, GVHD was difficult to distinguish from cytomegalovirus disease or drug reactions. The diagnosis was confirmed by demonstration of substantial donor lymphoid chimerism. METHODS: To identify risk factors for severe GVHD, a retrospective analysis was performed comparing index cases with the rest of the cases in our institutional experience. RESULTS: Closely matched human leukocyte antigen recipients, those older than 65 years, and recipients with donors more than 40 years younger were at higher risk for GVHD. One case occurred in a patient with a congenital immunodeficiency. CONCLUSIONS: Liver transplant-associated GVHD is a progressive and fatal disease. Future approaches should focus on prevention and might include avoidance of closely matched human leukocyte antigen donors, treatment of the donor to reduce the number of lymphocytes, or reduction of immunosuppression in the early posttransplant period.


Sujet(s)
Maladie du greffon contre l'hôte/étiologie , Transplantation hépatique/effets indésirables , Adolescent , Adulte , Facteurs âges , Sujet âgé , Enfant , Enfant d'âge préscolaire , Femelle , Maladie du greffon contre l'hôte/diagnostic , Maladie du greffon contre l'hôte/thérapie , Test d'histocompatibilité , Humains , Déficits immunitaires/congénital , Mâle , Adulte d'âge moyen , Études rétrospectives , Facteurs de risque
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...