Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
3.
Bioorg Med Chem ; 28(2): 115247, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31843461

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory disease that causes severe joints damage and other extra-articular alterations. Despite the efficacy of low-dose methotrexate (LD-MTX) in RA treatment, adverse effects are the predominant reasons for discontinuation of therapy. As a therapeutic targeting strategy, the presence of increased concentrations of reactive oxygen species (ROS) in the inflammatory environment can serve as the stimulus for prodrug activation in site-selective drug delivery systems. Our group has previously reported novel ROS sensitive prodrugs (1-3) of MTX and aminopterin (AMT) for site-selective delivery to inflammatory tissue associated with RA, with the aim of reducing side effects in RA therapy. Herein, we investigate the effect and toxicity of the same prodrugs in a rat CIA (collagen-induced arthritis) model of RA. We find that prodrug 1, an arylboronic acid ROS-sensitive MTX-prodrug, displays similar in vivo efficacy as MTX at an equimolar dose, while avoiding adverse effects known to restrict MTX treatment. To further characterize prodrug 1 and its ROS mediated activation, we synthesized compound 4, a negative control lacking the boronic acid moiety. We then investigated the effect of molecules on cell proliferation and cytotoxicity in the presence of the ROS scavenger pyruvate, as well as their stability in buffer and cell media, demonstrating a direct correlation between ROS concentration and the prodrug activity. Moreover, the in vitro ADME properties were investigated, including permeability, rat plasma and microsomal stability.


Subject(s)
Aminopterin/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Methotrexate/pharmacology , Prodrugs/pharmacology , Aminopterin/administration & dosage , Aminopterin/chemistry , Animals , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/chemistry , Apoptosis/drug effects , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/metabolism , Injections, Intraperitoneal , Methotrexate/administration & dosage , Methotrexate/chemistry , Molecular Structure , Prodrugs/administration & dosage , Prodrugs/chemistry , Rats , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
5.
Haematologica ; 104(5): 993-1003, 2019 05.
Article in English | MEDLINE | ID: mdl-30573506

ABSTRACT

Mogamulizumab, a humanized defucosylated anti-C-C chemokine receptor 4 monoclonal antibody, has been approved in Japan for the treatment of C-C chemokine receptor 4-positive adult T-cell leukemia/lymphoma (ATL). This phase II study evaluated efficacy and safety of mogamulizumab in ATL patients with acute, lymphoma, and chronic subtypes with relapsed/refractory, aggressive disease in the US, Europe, and Latin America. With stratification by subtype, patients were randomized 2:1 to intravenous mogamulizumab 1.0 mg/kg once weekly for 4 weeks and biweekly thereafter (n=47) or investigator's choice of chemotherapy (n=24). The primary end point was confirmed overall response rate (cORR) confirmed on a subsequent assessment at 8 weeks by blinded independent review. ORR was 11% (95%CI: 4-23%) and 0% (95%CI: 0-14%) in the mogamulizumab and chemotherapy arms, respectively. Best response was 28% and 8% in the respective arms. The observed hazard ratio for progression-free survival was 0.71 (95%CI: 0.41-1.21) and, after post hoc adjustment for performance status imbalance, 0.57 (95%CI: 0.337-0.983). The most frequent treatment-related adverse (grade ≥3) events with mogamulizumab were infusion-related reaction and thrombocytopenia (each 9%). Relapsed/refractory ATL is an aggressive, poor prognosis disease with a high unmet need. Investigator's choice chemotherapy did not result in tumor response in this trial; however, mogamulizumab treatment resulted in 11% cORR, with a tolerable safety profile.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Neoplasm Recurrence, Local/drug therapy , Salvage Therapy , Adult , Aged , Aged, 80 and over , Aminopterin/administration & dosage , Aminopterin/analogs & derivatives , Antibodies, Monoclonal, Humanized/administration & dosage , Cisplatin/administration & dosage , Cytarabine/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dexamethasone/administration & dosage , Female , Follow-Up Studies , Humans , International Agencies , Leukemia-Lymphoma, Adult T-Cell/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Oxaliplatin/administration & dosage , Prognosis , Retrospective Studies , Survival Rate , Young Adult , Gemcitabine
6.
Sci Rep ; 9(1): 20302, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31889144

ABSTRACT

Peripheral T-cell lymphomas (PTCL) are a heterogeneous group of non-Hodgkin's lymphomas with poor clinical outcomes. Pralatrexate showed efficacy and safety in recurrent or refractory PTCLs. The purpose or this study was to investigate the efficacy and safety of pralatrexate in relapsed or refractory PTCLs in real-world practice. This was an observational, multicenter, retrospective analysis. Between December 2012 and December 2016, a total of 38 patients with relapsed or refractory PTCLs were treated with pralatrexate at 10 tertiary hospitals in Korea. Patients received an intravenous infusion of pralatrexate at a dose of 30 mg/m2/week for 6 weeks on a 7-week schedule. Modified dosing and/or scheduling was allowed according to institutional protocols. Median patient age was 58 years (range, 29-80 years) and the most common subtype was peripheral T-cell lymphoma, not otherwise specified (n = 23, 60.5%). The median dosage of pralatrexate per administration was 25.6 mg/m2/wk (range, 15.0-33.0 mg/m2/wk). In intention-to-treat analysis, 3 patients (7.9%) showed a complete response and 5 patients (13.2%) showed a partial response, resulting in an overall response rate (ORR) of 21.1%. The median duration of response was 7.6 months (range, 1.6-24.3 months). The median progression-free survival (PFS) was 1.8 months (95% confidence interval [CI], 1.7-1.8 months) and the median overall survival was 7.7 months (95% CI, 4.4-9.0 months). The most common grade 3/4 adverse events were thrombocytopenia (n = 13, 34.2%), neutropenia (n = 7, 23.7%), and anemia (n = 7, 18.4%). Our study showed relatively lower ORR and shorter PFS in patients with recurrent or refractory PTCLs treated with pralatrexate in real-world practice. The toxicity profile was acceptable and manageable. We also observed significantly lower dose intensity of pralatrexate in real-world practice.


Subject(s)
Aminopterin/analogs & derivatives , Antineoplastic Agents/therapeutic use , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphoma, T-Cell, Peripheral/pathology , Aged , Aged, 80 and over , Aminopterin/administration & dosage , Aminopterin/adverse effects , Aminopterin/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Disease Management , Drug Resistance, Neoplasm , Female , Humans , Lymphoma, T-Cell, Peripheral/mortality , Male , Middle Aged , Neoplasm Staging , Prognosis , Recurrence , Retreatment , Retrospective Studies , Treatment Outcome
7.
Curr Opin Oncol ; 30(5): 285-291, 2018 09.
Article in English | MEDLINE | ID: mdl-30096094

ABSTRACT

PURPOSE OF REVIEW: Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of mature T-cell and natural killer (NK)-cell neoplasms in the WHO 2016 classification. Patient prognosis is poor when treated with CHOP, and there is an unmet need for new drugs. Several agents have been developed for PTCL, and their use is the subject of this review. RECENT FINDINGS: Phase 2 studies demonstrated the activity of new drugs in Relapsed/refractory PTCL. Only four compounds were approved by the food and drug administration: romidepsin and belinostat, which are epigenetic modifiers, the antifolate agent pralatrexate, the immuno-conjugate brentuximab vedotin. New combinations have been tested, but the results were disappointing. Given the latest progress in biology, targeted agents are evaluated in different subtypes of PTCL. Relapsed anaplastic large-cell lymphoma exhibits improved prognosis with the approved anti-CD30 drug conjugate brentuximab vedotin. Localized nasal NK/T is treated with radiotherapy and nonanthracycline chemotherapy with L-asparaginase. Recently, immune checkpoint inhibitors demonstrated activity in NK/T lymphoma and can be used in elderly patients. SUMMARY: Treatment remains a challenge for PTCL, and several targeted drugs provide new approaches. Progress will be made incrementally in the different subtypes. One of the critical situations facing new drugs is the ability to run robust clinical trials in rare diseases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, T-Cell, Peripheral/therapy , Aminopterin/administration & dosage , Aminopterin/analogs & derivatives , Brentuximab Vedotin , Clinical Trials, Phase II as Topic , Depsipeptides/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Immunoconjugates/administration & dosage , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphoma, T-Cell, Peripheral/immunology , Lymphoma, T-Cell, Peripheral/radiotherapy , Randomized Controlled Trials as Topic , Sulfonamides/administration & dosage
8.
Blood ; 131(4): 397-407, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29141948

ABSTRACT

Peripheral T-cell lymphomas (PTCL) are a group of rare malignancies characterized by chemotherapy resistance and poor prognosis. Romidepsin and pralatrexate were approved by the US Food and Drug Administration for patients with relapsed/refractory PTCL, exhibiting response rates of 25% and 29% respectively. Based on synergy in preclinical models of PTCL, we initiated a phase 1 study of pralatrexate plus romidepsin in patients with relapsed/refractory lymphoma. This was a single institution dose-escalation study of pralatrexate plus romidepsin designed to determine the dose-limiting toxicities (DLTs), maximum tolerated dose, pharmacokinetic profile, and response rates. Patients were treated with pralatrexate (10 to 25 mg/m2) and romidepsin (12 to 14 mg/m2) on 1 of 3 schedules: every week × 3 every 28 days, every week × 2 every 21 days, and every other week every 28 days. Treatment continued until progression, withdrawal of consent, or medical necessity. Twenty-nine patients were enrolled and evaluable for toxicity. Coadministration of pralatrexate and romidepsin was safe, well tolerated, with 3 DLTs across all schedules (grade 3 oral mucositis × 2; grade 4 sepsis × 1). The recommended phase 2 dose was defined as pralatrexate 25 mg/m2 and romidepsin 12 mg/m2 every other week. Twenty-three patients were evaluable for response. The overall response rate was 57% (13/23) across all patients and 71% (10/14) in PTCL. The phase 1 study of pralatrexate plus romidepsin resulted in a high response rate in patients with previously treated PTCL. A phase 2 study in PTCL will determine the efficacy of the combination. This trial was registered at www.clinicaltrials.gov as #NCT01947140.


Subject(s)
Aminopterin/analogs & derivatives , Antibiotics, Antineoplastic/therapeutic use , Depsipeptides/therapeutic use , Folic Acid Antagonists/therapeutic use , Lymphoma, T-Cell/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Aminopterin/administration & dosage , Aminopterin/adverse effects , Aminopterin/blood , Aminopterin/therapeutic use , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/blood , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Depsipeptides/administration & dosage , Depsipeptides/adverse effects , Depsipeptides/blood , Female , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/adverse effects , Folic Acid Antagonists/blood , Humans , Male , Middle Aged , Young Adult
9.
Cancer Sci ; 108(10): 2061-2068, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28771889

ABSTRACT

Pralatrexate is a novel antifolate approved in the USA for the treatment of relapsed or refractory peripheral T-cell lymphoma. To assess its safety, efficacy, and pharmacokinetics in Japanese patients with this disease, we undertook a phase I/II study. Pralatrexate was given i.v. weekly for 6 weeks of a 7-week cycle. All patients received concurrent vitamin B12 and folic acid. In phase I, three patients received pralatrexate 30 mg/m2 and none experienced a dose-limiting toxicity. In phase II, we treated 22 additional patients with that dose. The median number of treatment cycles was 1 (range, 1-9). Nine of 20 evaluable patients (45%) achieved an objective response by central review, including two complete responses. All responses occurred within the first treatment cycle. At the time of data cut-off, median progression-free survival was 150 days. Median overall survival was not reached. In the total population, the most commonly reported adverse events included mucositis (88%), thrombocytopenia (68%), liver function test abnormality (64%), anemia (60%), and lymphopenia (56%). Grade 3/4 adverse events included lymphopenia (52%), thrombocytopenia (40%), leukopenia (28%), neutropenia (24%), anemia (20%), and mucositis (20%). The pharmacokinetic profile showed no drug accumulation with repeat dosing. These results indicate that pralatrexate is generally well tolerated and effective in Japanese patients with relapsed or refractory peripheral T-cell lymphoma. This trial was registered with ClinicalTrials.gov (NCT02013362).


Subject(s)
Aminopterin/analogs & derivatives , Folic Acid/administration & dosage , Lymphoma, T-Cell, Peripheral/drug therapy , Neoplasm Recurrence, Local/drug therapy , Vitamin B 12/administration & dosage , Adult , Aged , Aged, 80 and over , Aminopterin/administration & dosage , Aminopterin/adverse effects , Aminopterin/pharmacokinetics , Drug Administration Schedule , Female , Folic Acid/therapeutic use , Humans , Japan , Male , Middle Aged , Survival Analysis , Treatment Outcome , Vitamin B 12/therapeutic use
10.
Clin Cancer Res ; 23(14): 3552-3556, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28167509

ABSTRACT

Purpose: Pralatrexate is a folic acid analogue metabolic inhibitor similar to methotrexate, which has shown tolerability and efficacy with an overall response rate of 45% in a phase I dose deescalation study of patients with relapsed/refractory cutaneous T-cell lymphoma (CTCL).Experimental Design: The object of this phase I/II open-label, multicenter clinical trial was to determine the MTD and recommended dose of pralatrexate plus oral bexarotene in 34 patients with relapsed/refractory CTCL who had failed prior systemic therapies. Pralatrexate was administered by intravenous push at 15 mg/m2 given weekly 3 weeks out of 4 weeks with daily oral bexarotene (150 or 300 mg/m2), levothyroxine, atorvastatin, folate, and with B12 every 2 months.Results: At the MTD of 15 mg/m2 bexarotene and 15 mg/m2 pralatrexate, the response rate was 60% [4 complete responses (CR), 14 partial responses (PR)], the maximum observed response duration was 28.9+ months, and duration of response for 4 CRs ranged from 9.0 to 28.3 months. The median progression-free survival was 12.8 months (0.5-29.9). Mucositis was the most common adverse event.Conclusions: The combination of pralatrexate (15 mg/m2) and oral bexarotene (150 mg/m2) is active with high response rates and minimal toxicity for cutaneous T-cell lymphomas. Clin Cancer Res; 23(14); 3552-6. ©2017 AACR.


Subject(s)
Aminopterin/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Lymphoma, T-Cell, Cutaneous/drug therapy , Tetrahydronaphthalenes/administration & dosage , Adult , Aged , Aged, 80 and over , Aminopterin/administration & dosage , Aminopterin/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bexarotene , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Lymphoma, T-Cell, Cutaneous/epidemiology , Lymphoma, T-Cell, Cutaneous/pathology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Tetrahydronaphthalenes/adverse effects , Treatment Outcome
11.
Cancer ; 122(21): 3297-3306, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27421044

ABSTRACT

BACKGROUND: The objective of this phase 1 and 2 trial was to identify the appropriate dose of combined carboplatin and pralatrexate for patients with recurrent, platinum-sensitive ovarian, fallopian tube, and primary peritoneal cancer. METHODS: In phase 1, patients received carboplatin (at an area under the curve of 5) and increasing doses of pralatrexate until the maximum-tolerated dose (MTD) of pralatrexate was achieved. The primary endpoint was the response rate. Additional endpoints were safety, response duration, progression-free survival, overall survival, and pharmacokinetics. RESULTS: Thirty patients were enrolled in phase 1, and 20 were enrolled in phase 2. Of all 50 patients, 49 completed the study. The mean patient age was 59 years, and patients completed a median of 6 cycles. The MTD for pralatrexate was 105 mg/m2 . The clinical benefit rate (complete responses plus partial responses plus stable disease) was 86%. Of 26 patients who received the MTD, 12 had a partial response, 11 had stable disease, and 2 had disease progression. The progression-free survival rate at 3 and 6 months was 87% and 79%, respectively; and the overall survival rate was 98% at 6 and 12 months and 66% at 24 months. Of 30 patients, 18 (60%) in phase 1 experienced an adverse event of any grade; and, of those, 4 patients (13%) had a grade 3 or greater adverse event. In phase 2, 12 patients (60%) had an adverse event of any grade, and 4 (20%) had grade 3 or greater toxicity. There was a significant reduction in the total body clearance of pralatrexate when it was received concurrently with carboplatin. CONCLUSIONS: Most patients responded to carboplatin-pralatrexate combination. This regimen is well tolerated and effective in this patient population. Cancer 2016;122:3297-3306. © 2016 American Cancer Society.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fallopian Tube Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Adult , Aged , Aminopterin/administration & dosage , Aminopterin/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carboplatin/administration & dosage , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/pathology , Carcinosarcoma/drug therapy , Carcinosarcoma/pathology , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/pathology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Fallopian Tube Neoplasms/pathology , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Prognosis , Survival Rate , Tissue Distribution
12.
Br J Haematol ; 173(2): 253-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27040320

ABSTRACT

Pralatrexate inhibits folic acid metabolism, and preclinical studies have shown that it is cytotoxic to multiple myeloma cells. This phase 1 study investigated the safety and efficacy of pralatrexate in combination with bortezomib in adults with relapsed or refractory multiple myeloma. A standard 3 + 3 design was used. Patients received intravenous pralatrexate at doses ranging from 10 to 30 mg/m(2) and intravenous bortezomib at a dose of 1·3 mg/m(2) on days 1, 8 and 15 of each 4-week cycle. Eleven patients were enrolled and completed a median of two cycles. The maximum tolerated dose was 20 mg/m(2) . Two patients experienced dose-limiting toxicity of mucositis. The most frequent non-haematological toxicities were fatigue (55%) and mucositis (45%). There were three serious adverse events in three patients: rash, sepsis and hypotension. One patient (9%) had a very good partial response, 1 (9%) had a partial response, 1 (9%) had minimal response and two (18%) had progressive disease. The median duration of response was 4 months, the median time to next treatment was 3·4 months and the median time to progression was 4 months. Pralatrexate, in combination with bortezomib, was generally safe and demonstrated modest activity in relapsed or refractory multiple myeloma. Clinicaltrials.gov identifier: NCT01114282.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Aged , Aminopterin/administration & dosage , Aminopterin/adverse effects , Aminopterin/analogs & derivatives , Bortezomib/administration & dosage , Bortezomib/adverse effects , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Treatment Outcome
13.
Pharmacotherapy ; 36(2): e8-e11, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26809959

ABSTRACT

Pralatrexate (PDX) is a folate antagonist structurally similar to methotrexate (MTX). Unlike MTX, it is currently not known whether PDX exhibits delayed clearance and heightened toxicity in the setting of fluid overload. A specific serum assay for PDX is not commercially available. To our knowledge, we report the first case using an MTX serum assay as a surrogate for PDX concentrations to avoid a potential drug-drug interaction with pralatrexate. We describe a 76-year-old man with refractory cutaneous T-cell lymphoma who began therapy with weekly PDX 15 mg/m(2) intravenous infusions on days 1, 8, and 15 of a 28-day cycle. He subsequently developed mucositis, a moderate right-sided pleural effusion, and peripheral edema over the next 5 weeks. Aggressive diuresis with furosemide was initiated, which was then withheld the day before his next PDX dose to avoid a potential drug-drug interaction between PDX and furosemide. His baseline MTX/PDX concentration (measured prior to administration of the cycle 2, week 2 PDX dose) was less than 0.20 µmol/L (i.e., undetectable). After PDX administration, his 1-hour peak MTX/PDX concentration increased to 0.58 µmol/L. Aggressive diuresis was withheld until his MTX/PDX concentration was undetectable, 43.5 hours later. PDX is more potent than MTX and displays similar pharmacokinetic properties. PDX concentrations using the serum MTX assay reflect lower values than those reported from PDX-specific assays in clinical studies. Because PDX is approved by the U.S. Food and Drug Administration for the treatment of uncommon malignancies, it is unlikely that a specific assay will be commercially developed. We propose that the MTX serum assay has merit for use in determining when to reinstate possible interacting drug therapies such as loop diuretics.


Subject(s)
Aminopterin/analogs & derivatives , Folic Acid Antagonists/blood , Lymphoma, T-Cell, Cutaneous/blood , Skin Neoplasms/blood , Aged , Aminopterin/administration & dosage , Aminopterin/blood , Aminopterin/pharmacokinetics , Aminopterin/therapeutic use , Drug Interactions , Drug Monitoring , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/pharmacokinetics , Folic Acid Antagonists/therapeutic use , Furosemide/administration & dosage , Furosemide/adverse effects , Furosemide/therapeutic use , Humans , Infusions, Intravenous , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/physiopathology , Male , Methotrexate/analysis , Methotrexate/chemistry , Pleural Effusion/drug therapy , Pleural Effusion/etiology , Reagent Kits, Diagnostic , Skin Neoplasms/complications , Skin Neoplasms/drug therapy , Skin Neoplasms/physiopathology , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/adverse effects , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use , Treatment Outcome
14.
Br J Haematol ; 172(4): 535-44, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26627450

ABSTRACT

Peripheral T-cell lymphomas (PTCL) have suboptimal outcomes using conventional CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone) chemotherapy. The anti-folate pralatrexate, the first drug approved for patients with relapsed/refractory PTCL, provided a rationale to incorporate it into the front-line setting. This phase 2 study evaluated a novel front-line combination whereby cyclophosphamide, etoposide, vincristine and prednisone (CEOP) alternated with pralatrexate (CEOP-P) in PTCL. Patients achieving a complete or partial remission (CR/PR) were eligible for consolidative stem cell transplantation (SCT) after 4 cycles. Thirty-three stage II-IV PTCL patients were treated: 21 PTCL-not otherwise specified (64%), 8 angioimmunoblastic T cell lymphoma (24%) and 4 anaplastic large cell lymphoma (12%). The majority (61%) had stage IV disease and 46% were International Prognostic Index high/intermediate or high risk. Grade 3-4 toxicities included anaemia (27%), thrombocytopenia (12%), febrile neutropenia (18%), mucositis (18%), sepsis (15%), increased creatinine (12%) and liver transaminases (12%). Seventeen patients (52%) achieved a CR. The 2-year progression-free survival and overall survival, were 39% (95% confidence interval 21-57) and 60% (95% confidence interval 39-76), respectively. Fifteen patients (45%) (12 CR) received SCT and all remained in CR at a median follow-up of 21·5 months. CEOP-P did not improve outcomes compared to historical data using CHOP. Defining optimal front line therapy in PTCL continues to be a challenge and an unmet need.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, T-Cell, Peripheral/drug therapy , Adult , Aged , Aged, 80 and over , Aminopterin/administration & dosage , Aminopterin/adverse effects , Aminopterin/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Disease Progression , Disease-Free Survival , Epirubicin/administration & dosage , Epirubicin/adverse effects , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prednisone/administration & dosage , Prednisone/adverse effects , Treatment Outcome , Vincristine/administration & dosage , Vincristine/adverse effects
15.
Cancer ; 121(21): 3862-8, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26242208

ABSTRACT

BACKGROUND: Pralatrexate (PDX) is an inhibitor of dihydrofolate reductase that was rationally designed to improve cellular uptake and retention of the drug. Preclinical data have shown synergy with the sequential administration of a dihydrofolate reductase inhibitor followed 24 hours later by 5-fluorouracil (5-FU). METHODS: Twenty-seven patients were enrolled at 1 of 5 PDX dose levels from 75 to 185 mg/m(2) on day 1 followed 24 hours later by 5-FU at a dose of 3000 mg/m(2) /48 hours every 2 weeks with folic acid and vitamin B12 supplementation. Baseline blood was collected for pharmacogenetic analysis of polymorphisms of methylenetetrahydrofolate reductase and thymidylate synthase. RESULTS: Mucositis was the most common dose-limiting toxicity. When the worst toxicities across all cycles were considered, grade 3 to 4 neutropenia, anemia, and thrombocytopenia were found to have occurred in 14.8%, 14.8%, and 0% of patients, respectively. Grade 2 to 3 toxicities included mucositis (66.6%), dehydration (33.3%), fatigue (25.9%), and diarrhea (22.2%). Version 3.0 of the National Cancer Institute Common Toxicity Criteria was used to grade toxicities The median progression-free survival (PFS) was 112 days (range, 28-588 days). Seven patients (26%) had a PFS of >180 days (5 patients with colorectal cancer, 1 patient with pancreatic cancer, and 1 patient with non-small cell lung cancer). Polymorphisms in methylenetetrahydrofolate reductase and thymidylate synthase did not correlate with toxicity. CONCLUSIONS: The recommended dose of PDX was 148 mg/m(2) . A subset of heavily pretreated patients had PFS durations of ≥6 months with this regimen.


Subject(s)
Aminopterin/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/drug therapy , Colorectal Neoplasms/drug therapy , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Gastrointestinal Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Aminopterin/administration & dosage , Aminopterin/adverse effects , Aminopterin/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Female , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/adverse effects , Folic Acid Antagonists/therapeutic use , Humans , Incidence , Male , Middle Aged , Mucositis/chemically induced , Mucositis/epidemiology , Polymorphism, Genetic/genetics , Tetrahydrofolate Dehydrogenase/blood , Tetrahydrofolate Dehydrogenase/genetics , Thymidylate Synthase/blood , Thymidylate Synthase/genetics , Time Factors , Treatment Outcome
16.
Clin Cancer Res ; 21(18): 4097-109, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25878331

ABSTRACT

PURPOSE: Aurora A kinase (AAK) is expressed exclusively during mitosis, and plays a critical role in centrosome duplication and spindle formation. Alisertib is a highly selective AAK inhibitor that has demonstrated marked clinical activity of alisertib across a spectrum of lymphomas, though particularly in patients with T-cell lymphoma (TCL). We sought to compare and contrast the activity of alisertib in preclinical models of B-cell lymphoma (BCL) and TCL, and identify combinations worthy of clinical study. High-throughput screening of pralatrexate, the proteasome inhibitor (ixazomib), and the histone deacetylase (HDAC) inhibitor (romidepsin) revealed that only romidepsin synergized with alisertib, and only in models of TCL. We discovered that the mechanism of synergy between AAK inhibitors and HDAC inhibitors appears to be mediated through cytokinesis failure. EXPERIMENTAL DESIGN: A high-throughput screening approach was used to identify drugs that were potentially synergistic in combination with alisertib. Live-cell imaging was used to explore the mechanistic basis for the drug: drug interaction between alisertib and romidepsin. An in vivo xenograft TCL model was used to confirm in vitro results. RESULTS: In vitro, alisertib exhibited concentration-dependent cytotoxicity in BCL and TCL cell lines. Alisertib was synergistic with romidepsin in a T-cell-specific fashion that was confirmed in vivo. Live-cell imaging demonstrated that the combination treatment resulted in profound cytokinesis failure. CONCLUSIONS: These data strongly suggest that the combination of alisertib and romidepsin is highly synergistic in TCL through modulation of cytokinesis and merits clinical development.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Histone Deacetylase Inhibitors/chemistry , Lymphoma, T-Cell/immunology , Protein Kinase Inhibitors/chemistry , Aminopterin/administration & dosage , Aminopterin/analogs & derivatives , Animals , Aurora Kinase A/metabolism , Azepines/administration & dosage , Azepines/therapeutic use , Boron Compounds/administration & dosage , Cell Cycle , Cell Line, Tumor , Centrosome/ultrastructure , Cytokinesis , Depsipeptides/administration & dosage , Drug Synergism , Glycine/administration & dosage , Glycine/analogs & derivatives , Histone Deacetylases/metabolism , Humans , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Lymphoma, T-Cell/drug therapy , Mice , Mice, SCID , Mitosis , Neoplasm Transplantation , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Spindle Apparatus , Xenograft Model Antitumor Assays
17.
Clin Cancer Res ; 21(9): 2096-106, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25677697

ABSTRACT

PURPOSE: T-cell lymphomas (TCL) are aggressive diseases, which carry a poor prognosis. The emergence of new drugs for TCL has created a need to survey these agents in a rapid and reproducible fashion, to prioritize combinations which should be prioritized for clinical study. Mouse models of TCL that can be used for screening novel agents and their combinations are lacking. Developments in noninvasive imaging modalities, such as surface bioluminescence (SBL) and three-dimensional ultrasound (3D-US), are challenging conventional approaches in xenograft modeling relying on caliper measurements. The recent approval of pralatrexate and romidepsin creates an obvious combination that could produce meaningful activity in TCL, which is yet to be studied in combination. EXPERIMENTAL DESIGN: High-throughput screening and multimodality imaging approach of SBL and 3D-US in a xenograft NOG mouse model of TCL were used to explore the in vitro and in vivo activity of pralatrexate and romidepsin in combination. Corresponding mass spectrometry-based pharmacokinetic and immunohistochemistry-based pharmacodynamic analyses of xenograft tumors were performed to better understand a mechanistic basis for the drug:drug interaction. RESULTS: In vitro, pralatrexate and romidepsin exhibited concentration-dependent synergism in combination against a panel of TCL cell lines. In a NOG murine model of TCL, the combination of pralatrexate and romidepsin exhibited enhanced efficacy compared with either drug alone across a spectrum of tumors using complementary imaging modalities, such as SBL and 3D-US. CONCLUSIONS: Collectively, these data strongly suggest that the combination of pralatrexate and romidepsin merits clinical study in patients with TCLs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Disease Models, Animal , Lymphoma, T-Cell/drug therapy , Xenograft Model Antitumor Assays , Aminopterin/administration & dosage , Aminopterin/analogs & derivatives , Animals , Cell Line, Tumor , Depsipeptides/administration & dosage , Drug Synergism , Flow Cytometry , Humans , Luminescent Measurements , Mice , Transfection
18.
PLoS One ; 9(9): e108303, 2014.
Article in English | MEDLINE | ID: mdl-25255447

ABSTRACT

BACKGROUND: Options are limited for patients with atopic dermatitis (AD) who do not respond to topical treatments. Antifolate therapy with systemic methotrexate improves the disease, but is associated with adverse effects. The investigational antifolate LD-aminopterin may offer improved safety. It is not known how antifolate dose and dosing frequency affect efficacy in AD, but a primary mechanism is thought to involve the antifolate-mediated accumulation of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR). However, recent in vitro studies indicate that AICAR increases then decreases as a function of antifolate concentration. To address this issue and understand how dosing affects antifolate efficacy in AD, we examined the efficacy and safety of different oral doses and schedules of LD-aminopterin in the canine model of AD. METHODS AND FINDINGS: This was a multi-center, double-blind trial involving 75 subjects with canine AD randomized to receive up to 12 weeks of placebo, once-weekly (0.007, 0.014, 0.021 mg/kg) or twice-weekly (0.007 mg/kg) LD-aminopterin. The primary efficacy outcome was the Global Score (GS), a composite of validated measures of disease severity and itch. GS improved in all once-weekly cohorts, with 0.014 mg/kg being optimal and significant (43%, P<0.01). The majority of improvement was seen by 8 weeks. In contrast, GS in the twice-weekly cohort was similar to placebo and worse than all once-weekly cohorts. Adverse events were similar across all treated cohorts and placebo. CONCLUSIONS: Once-weekly LD-aminopterin was safe and efficacious in canine AD. Twice-weekly dosing negated efficacy despite having the same daily and weekly dose as effective once-weekly regimens. Optimal dosing in this homologue of human AD correlated with the concentration-selective accumulation of AICAR in vitro, consistent with AICAR mediating LD-aminopterin efficacy in AD.


Subject(s)
Aminopterin/pharmacology , Dermatitis, Atopic/drug therapy , Folic Acid Antagonists/pharmacology , Administration, Oral , Aminopterin/administration & dosage , Animals , Dogs , Drug Administration Schedule , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/adverse effects , Humans , Prednisone/administration & dosage , Treatment Outcome
19.
Cancer Chemother Pharmacol ; 74(5): 1029-32, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25205429

ABSTRACT

PURPOSE: To investigate the ability of leucovorin (LV) to abrogate dose-limiting toxicities of pralatrexate (PDX) while maintaining efficacy, in vivo. METHODS: H2052 mesothelioma cells were treated with the antifolates methotrexate (MTX), PDX and pemetrexed, with and without LV rescue 24 h later. Cell killing was evaluated 48 h later. Female nude mice bearing H2052 xenografts were treated with varying doses and schedules of the antifolate PDX and LV. RESULTS: In vitro, H2052 cells were more sensitive to PDX as compared to MTX and pemetrexed. Administration of LV 24 h after antifolate treatment reduced efficacy of antifolates MTX and pemetrexed, but not PDX. In vivo, LV was found to reduce toxicity of PDX at the maximum tolerated dose without sacrificing efficacy. Lethal doses of PDX were rescued by LV, and mice bearing the H2052 tumor demonstrated prolonged and enhanced tumor regression. CONCLUSIONS: High-dose PDX with subsequent LV rescue may be a viable treatment strategy in mesothelioma and other cancers. The inclusion of LV rescue into new and existing PDX treatment protocols should be explored as a way to expand the tolerability and effectiveness of PDX in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mesothelioma/drug therapy , Xenograft Model Antitumor Assays , Aminopterin/administration & dosage , Aminopterin/analogs & derivatives , Aminopterin/pharmacology , Animals , Body Weight/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/pharmacology , Glutamates/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Leucovorin/administration & dosage , Leucovorin/pharmacology , Mesothelioma/pathology , Methotrexate/pharmacology , Mice, Nude , Pemetrexed , Time Factors , Tumor Burden/drug effects , Vitamin B Complex/administration & dosage , Vitamin B Complex/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...