Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Clin Genitourin Cancer ; 22(3): 102054, 2024 Jun.
Article En | MEDLINE | ID: mdl-38457853

INTRODUCTION: Evidence is limited on whether fibroblast growth factor receptor gene alterations (FGFRalt) impact clinical outcomes in patients with locally advanced or metastatic urothelial cancer (mUC). This study evaluated progression-free survival (PFS) in patients with mUC based on FGFRalt status in the first-line setting (1L). PATIENTS AND METHODS: Data on mUC patients were retrieved via convenience sampling of oncologists/urologists surveyed between August and September 2020 who treated at least 1 FGFRalt patient between July 2017 and June 2019. The questionnaire included information on patient demographics, FGFR status, treatment, and clinical and radiographic measures of progression. Primary endpoint was time from metastatic diagnosis to disease progression from initial treatment for FGFRalt and FGFRwt (wild-type) mUC. Cox proportional hazards models quantified adjusted risk of FGFR status relating to PFS. RESULTS: A total of 414 patients were analyzed. Mean age was 64.5 years, 73.9% were male, and 52.7% had an FGFRalt. Among FGFRalt, 47.2% received chemotherapy, 27.5% immune checkpoint inhibition (ICI), 11.5% chemotherapy+ICI, and 13.8% other treatments in 1L. FGFR status did not influence PFS from time of mUC diagnosis or among 224 stratified patients receiving either chemotherapy or chemotherapy+ICI. However, among 97 patients with an FGFRalt receiving 1L ICI therapy only, adjusted risk of progression was twice that of FGFRwt (HR: 2.12; 95% CI: 1.13-4.00). CONCLUSION: Although FGFRalt did not predict outcomes in the overall cohort, for patients treated with 1L ICI, FGFRalt had significantly higher rates of progression than FGFRwt patients. Further validation is needed to determine whether FGFRalt has a decreased benefit from ICI therapy.


Receptors, Fibroblast Growth Factor , Humans , Male , Female , Middle Aged , Aged , Prognosis , Receptors, Fibroblast Growth Factor/genetics , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/secondary , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/pathology , Progression-Free Survival , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/mortality , Mutation , Retrospective Studies , Immune Checkpoint Inhibitors/therapeutic use , Disease Progression
2.
Cancer Med ; 10(11): 3565-3574, 2021 06.
Article En | MEDLINE | ID: mdl-33960681

BACKGROUND: As with other alkylating agents, cardiac dysfunction can occur with trabectedin therapy for advanced soft tissue sarcomas (STS) or recurrent ovarian cancer (ROC) where treatment options for advanced disease are still limited. Cardiac safety for trabectedin monotherapy (T) for STS or in combination with pegylated liposomal doxorubicin (T+PLD) for ROC was evaluated in this retrospective postmarketing regulatory commitment. METHODS: Patient data for multiple cardiac-related treatment-emergent adverse events (cTEAEs) were evaluated in pooled analyses of ten phase 2 trials, one phase 3 trial in STS (n = 982), and two phase 3 trials in ROC (n = 1231). RESULTS: Multivariate analyses on pooled trabectedin data revealed that cardiovascular medical history (risk ratio [RR (95% CI)]: 1.90 [1.24-2.91]; p = 0.003) and age ≥65 years (RR [95% CI]: 1.78 [1.12-2.83]; p = 0.014) were associated with increased risk for cTEAEs. Multivariate analyses showed increased risk of experiencing cTEAEs with T+PLD compared to PLD monotherapy (RR [95% CI]: 2.70 [1.75-4.17]; p < 0.0001) and with history of prior cardiac medication (RR [95% CI]: 1.88 [1.16-3.05]; p = 0.010). CONCLUSIONS: For patients with STS or ROC who still have limited treatment options, trabectedin may be initiated after carefully considering benefit versus risk. Trial Registration (ClinicalTrials.gov): NCT01343277; NCT00113607; NCT01846611.


Antineoplastic Agents, Alkylating/adverse effects , Doxorubicin/analogs & derivatives , Heart/drug effects , Ovarian Neoplasms/drug therapy , Sarcoma/drug therapy , Trabectedin/adverse effects , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cardiotoxicity , Cardiovascular Diseases/diagnosis , Child , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/drug therapy , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Product Surveillance, Postmarketing , Retrospective Studies , Soft Tissue Neoplasms/drug therapy , Stroke Volume/drug effects , Trabectedin/administration & dosage , Ventricular Function, Left/drug effects , Young Adult
3.
Data Brief ; 30: 105465, 2020 Jun.
Article En | MEDLINE | ID: mdl-32346557

The data presented herein are supplementary to our published primary article "A phase 3 randomized, open-label, multicenter trial for safety and efficacy of combined trabectedin and pegylated liposomal doxorubicin therapy for recurrent ovarian cancer"[1]. The exploratory analysis evaluated the impact of prior pegylated liposomal doxorubicin (PLD) therapy in patients who participated in a randomized, open-label study comparing combination therapy of trabectedin and PLD vs PLD alone in third-line recurrent ovarian cancer (ROC). These exploratory analyses showed that prior treatment with PLD in ROC does not impact the response and survival rates nor does it increase toxicities or negatively influence survival and response rates in both treatment groups.

4.
Gynecol Oncol ; 156(3): 535-544, 2020 03.
Article En | MEDLINE | ID: mdl-31924332

OBJECTIVE: This phase 3 study aimed to compare overall survival (OS) of women with platinum-sensitive, recurrent ovarian cancer (ROC) treated with third-line trabectedin (T) + pegylated liposomal doxorubicin (PLD) vs. PLD monotherapy. METHODS: Women with advanced-relapsed epithelial ovarian cancer were randomly assigned 1: 1 to intravenous infusions of either T + PLD (trabectedin 1.1 mg/m2 for 3 h; PLD 30 mg/m2 for 1.5 h, every 3 weeks) or PLD (50 mg/m2 for 1.5 h, every 4 weeks). Primary endpoint was OS. Secondary endpoints included investigator-assessed progression free survival (PFS) and objective response rates (ORR). At randomization, patients were stratified by time from last dose of first-line platinum therapy to disease progression, ECOG grade 0 or 1, BRCA1/2 germline mutational status, and prior PLD therapy. Exploratory endpoints included OS, PFS, and ORR in the stratified subgroups (PFI, ECOG, BRCA1/2 status, and prior PLD therapy). This trial is registered with ClinicalTrials.gov, number NCT01846611. RESULTS: 576 patients were randomized (T + PLD, n = 289; PLD, n = 287). Median OS was 23.8 months with T + PLD vs. 22.2 months with PLD (HR:0.92, 95%CI:0.73-1.18; p = 0.52). Median PFS was 7.52 vs. 7.26 months (HR:0.93, 95%CI:0.76-1.15; p = 0.52); ORR was 46% vs. 35.9% (OR:1.52, 95%CI:1.07-2.16; p = 0.01). Patients with BRCA1/2 mutations had median OS of 34.2 months with T + PLD vs. 20.9 months with PLD (HR:0.54, 95%CI:0.33-0.90; p = 0.016). Patients with BRCA1/2 mutations had median PFS of 10.1 months with T + PLD vs. 7.6 months with PLD (HR:0.72, 95%CI:0.48-1.08; p = 0.039). Patients with BRCA1/2 mutations and a 6-12 months platinum-free interval (PFI), median OS was 31.5 vs. 14.9 months, respectively (HR:0.37, 95%CI:0.17-0.82; p = 0.011). Grade 3-4 AEs were higher in T + PLD (79%) vs. PLD (54%). CONCLUSION: Combination of T and PLD did not show favorable OS benefit nor safety; however, patients with germline BRCA1/2 mutations and/or a PFI of 6-12 months appear to have clinically relevant survival benefit with T + PLD. No new safety signals were identified.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/analogs & derivatives , Female , Humans , Infusions, Intravenous , Middle Aged , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Progression-Free Survival , Survival Rate , Trabectedin/administration & dosage , Trabectedin/adverse effects , Treatment Outcome , Young Adult
5.
Cancer ; 125(24): 4435-4441, 2019 Dec 15.
Article En | MEDLINE | ID: mdl-31503332

BACKGROUND: The results of the randomized, phase 3 ET743-SAR-3007 trial demonstrated that trabectedin had a significantly longer progression-free survival (PFS) compared with dacarbazine in patients with advanced leiomyosarcoma/liposarcoma after the failure of prior chemotherapy. Patients randomized to trabectedin received a 24-hour intravenous infusion either in an inpatient or outpatient setting. Herein, the authors reported the safety, efficacy, and patient-reported outcomes based on first infusion site of care. METHODS: Patients were randomized 2:1 to trabectedin (at a dose of 1.5 mg/m2 ) or dacarbazine (1 g/m2 over 20-120 minutes) with overall survival (OS) as the primary endpoint and PFS, time to disease progression, objective response rate, duration of response, safety, and patient-reported symptom scoring as secondary endpoints. The setting of the trabectedin infusion was based on institutional preference and categorized based on the setting of the first infusion. RESULTS: Of the 378 patients who were treated with trabectedin, 100 (27%) and 277 (73%), respectively, first received trabectedin in the inpatient and outpatient setting. No differences were observed with regard to PFS or OS based on site of care. The median PFS was 4.1 months versus 4.2 months (hazard ratio, 0.90; P = .49) for inpatients versus outpatients, respectively, and the median OS was 14.3 months versus 13.7 months (hazard ratio, 0.89; P = .40), respectively. Grade 3/4 adverse events (classified according to the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]) were reported in 87 inpatients (87%) compared with 219 outpatients (79%); grade 3/4 serious adverse events were reported in 43 inpatients (43%) and 92 outpatients (33%). Extravasation occurred in 0 inpatients and 5 outpatients (2%), whereas the incidence of catheter-related complications was similar between groups (16% vs 15%). CONCLUSIONS: Although the majority of patients who were randomized to trabectedin received outpatient therapy, the outcomes of the current study suggested equivalent safety and efficacy in either setting.


Antineoplastic Agents, Alkylating/administration & dosage , Inpatients , Leiomyosarcoma/drug therapy , Liposarcoma/drug therapy , Outpatients , Trabectedin/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/adverse effects , Disease Management , Female , Humans , Leiomyosarcoma/diagnosis , Leiomyosarcoma/mortality , Liposarcoma/diagnosis , Liposarcoma/mortality , Male , Middle Aged , Patient Reported Outcome Measures , Trabectedin/adverse effects , Treatment Outcome , Young Adult
6.
Med Devices (Auckl) ; 11: 241-252, 2018.
Article En | MEDLINE | ID: mdl-30100767

BACKGROUND: Current administration of hydroxyprogesterone caproate (HPC) by intramuscular injection is associated with limitations, including the potential for human error and contamination, patient anxiety, and increased risk of needlestick injury. OBJECTIVE: To describe the design of an auto-injector for subcutaneous (SC) administration of HPC and the results of studies that evaluated the target user's understanding of the proper use of this device. MATERIALS AND METHODS: A single-use, prefilled, fixed-dose, disposable auto-injector intended for the SC administration of HPC was developed, and its usability by health care providers was evaluated in 3 formative (N=32, 64 injections) and 3 validation studies (N=45, 90 injections). These studies consisted of one-on-one testing sessions performed in a simulated home environment. Analyses were based on observed use error or use difficulty during the performance of specific tasks, including those considered critical (associated with high severity harms). RESULTS: In the formative studies, the majority of participants correctly administered an injection with the auto-injector, but prior training improved performance. Specific errors were noted, including holding the device at the injection site for a period inconsistent with its instructions for use (IFU). The IFU was modified to reduce potential occurrence of these errors. Use errors were subsequently observed on critical tasks in the first and second validation studies, including hold-time errors that were attributed to using visual cues rather than counting seconds. For the third validation study, the IFU was modified to focus on visual cues and all users were able to successfully perform the injection per the IFU. CONCLUSION: An auto-injector device for SC administration of HPC for reduction in risk of recurrent preterm birth was successfully developed through iterative design and validation testing. The device design provides high usability and acceptance of this device by health care professionals.

7.
J Biomed Mater Res A ; 100(9): 2365-72, 2012 Sep.
Article En | MEDLINE | ID: mdl-22528373

Polymeric controlled delivery systems hold great promise in the field of modern medicine. Such technology has already been converted into commercially viable products in a myriad of fields. Chemotherapy is an example of such an area where constant efficacious levels of drug can greatly enhance clinical outcomes. The key to designing such therapies is the preparation of the proper delivery system. To this end, a series of bioresorbable polyether-ester-carbonate copolymers have been developed, which when combined with a diluent, are capable injection into the body and consistently forming a drug delivery depot. The study delineated here aimed at producing a more effective treatment of a common drug, paclitaxel, using the polymeric carrier. The polymer carrier system exhibited controlled release of paclitaxel both in vitro and in vivo. Drug concentrations were analyzed by high performance liquid chromatography and apoptotic activity was confirmed through flow cytometry. Relevant success was exhibited by the regression of tumor size following a multiple injection treatment regimen in a murine xenograft model. This multiple injection treatment shows promising results when compared to the traditional paclitaxel paradigm of a single injection for a period of 3 weeks.


Antineoplastic Agents, Phytogenic/administration & dosage , Carbonates/chemistry , Delayed-Action Preparations/chemistry , Paclitaxel/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Cell Line, Tumor , Female , Injections , Lung Neoplasms/drug therapy , Mice , Mice, Inbred C57BL , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use
8.
J Biomed Mater Res B Appl Biomater ; 69(2): 173-82, 2004 May 15.
Article En | MEDLINE | ID: mdl-15116407

An absorbable microparticulate cation exchanger was synthesized as a versatile carrier for biologically active proteins. In this work, acid-terminated polyglycolide (or polyglycolic acid) microparticulates (PG-MP) were surface modified for either sustained release of cytokines or as a platform for immunomodulation. The intended goal was to achieve in situ recruitment/maturation of dendritic cells and activation of T cells for tumor immunotherapy. PG-MP were prepared with a volume weighted mean diameter of 7.02 micro (range: 2.09-14.58 micro). Accessible carboxylic acid groups were determined to be 0.3 mmol/g with a corresponding zeta potential of -21.87 mV in phosphate-buffered saline. Under low magnification, scanning electron microscopy (SEM) revealed a highly textured surface due to processing from repetitive jet milling. However, a moderately porous architecture was noted at higher magnification. Electron spectroscopy for chemical analysis was used to characterize the PG-MP surface before and after adsorption of human granulocyte-macrophage colony stimulating factor (GM-CSF). Adsorption of GM-CSF on PG-MP (PG-GMCSF) resulted in a modest increase in the surface atomic concentration of nitrogen (0.97%). Pretreating the surface with poly-L-lysine (PG/Lys-GMCSF) prior to adding GM-CSF produced a nearly threefold increase in the surface nitrogen concentration (4.20% compared to 1.47%). This manipulation not only increased loading content, but also prolonged the release of GM-CSF released from 6 days to 26 days. ESCA on the post-release PG-MP samples (PG-GMCSF and PG/Lys-GMCSF) revealed a similar residual surface nitrogen concentration (2.26% vs. 2.35%). The observation was consistent with irreversibly adsorbed GM-CSF. It is postulated that irreversibly bound GM-CSF is released over time as a function of microparticulate degradation. Biological activity of released GM-CSF was confirmed by the proliferation of a GM-CSF-dependent cell line (TF-1) in the presence of microparticulates. PG-MP mediated activation of T cells was achieved through irreversible adsorption of either antimouse cd3 plus antimouse cd28 monoclonal antibodies (alpha-cd3/cd28-MP) or antihuman CD3 plus antihuman CD28 monoclonal antibodies (alpha-CD3/CD28-MP) on PG-MP. Irreversibly adsorbed antibodies were capable of activating both resting mouse and human T cells. Intracellular flow cytometry on mouse T cells revealed that nearly 50% of the activated cells produced interferon-gamma (IFN-gamma). This was consistent with a TH-1 or cell-mediated response. In vivo efficacy was evaluated in a mouse flank tumor model showing a significant antitumor effect both alone and in combination. Combination therapy was most effective at preventing tumor implantation (8/8 mice) and was able induce tumor regression (4/7 mice) and/or stable disease (3/7 mice) in a regression model. In these studies, immunohistochemistry was used to confirm local recruitment of dendritic cells. In conclusion, the PG-MP represents a novel absorbable cation exchanger that can be readily manipulated to deliver biologically active proteins for immunotherapy.


Drug Delivery Systems , Immunotherapy/methods , Ion Pumps/metabolism , Absorption , Animals , CD28 Antigens/metabolism , CD3 Complex/metabolism , Cell Division , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Ion Pumps/chemistry , Ion Pumps/therapeutic use , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/prevention & control , Neoplasms/therapy , Particle Size , Polyglycolic Acid/chemistry , Polyglycolic Acid/metabolism , Surface Properties , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
9.
J Control Release ; 91(1-2): 209-24, 2003 Aug 28.
Article En | MEDLINE | ID: mdl-12932653

Acid-terminated polyglycolide microparticles (PG-MP) were prepared as a versatile substrate that could be surface-modified for either immobilization of anti-cd3 and anti-cd28 mAb to activate T cells or sustained release of granulocyte-macrophage colony stimulating factor (GM-CSF) for dendritic cell (DC) recruitment and maturation. PG-MP were prepared with a volume-weighted mean diameter of 56 or 57 microm. Accessible carboxylic acid group concentration was determined by potentiometric titration to be 0.3 mmole/g and corresponded to a zeta potential of -21.87 mV. PG-MP immobilized with either anti-human CD3/CD28 or anti-mouse cd3/cd28 induced significant proliferation of T cells. Intracellular flow cytometry in activated mouse T cells was significant for IFN-gamma, but not IL-4. Microparticles surface-modified for GM-CSF release were prepared from either PG-MP or PG pre-treated with poly-L-lysine (PG-Lys) to manipulate surface charge. GM-CSF released from PG-Lys-MP was observed for up to 26 days. The biologic activity of released GM-CSF was confirmed by using a h-GM-CSF-dependent cell line. The efficacy of the alpha-cd3/cd28-MP and GMCSF-MP was studied in a syngeneic mouse tumor prevention and regression model. Co-injection of Meth A fibrosarcoma cells with alpha-cd3/cd28-MP and GMCSF-MP completely prevented tumor implantation (0/24). The regression model showed complete tumor regression in four of seven animals and stable disease in three of seven. In the latter study, a dramatic level of DC infiltration was observed compared to controls.


CD28 Antigens/immunology , CD3 Complex/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunotherapy , Neoplasms/therapy , Animals , Cytokines/chemistry , Dendritic Cells/immunology , Flow Cytometry , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Microscopy, Electron , Microspheres , Neoplasm Transplantation , Neoplasms/immunology , Neoplasms/prevention & control , Polyglycolic Acid/chemistry , T-Lymphocytes/immunology
10.
Clin Immunol ; 105(3): 259-72, 2002 Dec.
Article En | MEDLINE | ID: mdl-12498807

We compared the ability of two genetically modified myeloid cells, K562 and U937, to serve as artificial antigen-presenting cells (aAPC). Both aAPC were stably transfected with the low-affinity Fcgamma receptor CD32 (K32/U32 cells). K32 cells loaded with anti-CD3 and anti-CD28 Ab (K32/CD3/28) induced more rapid CD4 T-cell expansion than CD3/28-coated beads. In contrast, U32/CD3/28 induced high levels of CD4 T-cell thymidine uptake but were unable to sustain long-term T-cell expansion. K32 cells, but not U32 cells, loaded with anti-CD3 alone also stimulated CD4 T-cell growth and IL-2 secretion, indicating the expression of additional costimulatory molecules on K32 cells. We found constitutive expression of B7-H3 and a strong upregulation of mRNA encoding for IL-15, PD-L1, and PD-L2 after coculture with CD4 T cells activated by K32/CD3/28 but not U32/CD3/28. We conclude that K32 aAPCs are a robust system for clinical scale ex vivo expansion of CD4 T cells.


Antigen-Presenting Cells , Blood Proteins , CD28 Antigens/immunology , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , Peptides , Antibodies/immunology , Antigens, CD , B7 Antigens , B7-1 Antigen/genetics , B7-H1 Antigen , Cell Division , Cell Line , Coculture Techniques , DNA, Recombinant , Electroporation , Humans , Interleukin-15/genetics , Interleukin-2/metabolism , Lymphocyte Activation , Membrane Glycoproteins , RNA, Messenger , Receptors, Immunologic , Time Factors , Up-Regulation
...