Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Lancet ; 395(10236): 1547-1557, 2020 05 16.
Article in English | MEDLINE | ID: mdl-32416780

ABSTRACT

BACKGROUND: Atezolizumab can induce sustained responses in metastatic urothelial carcinoma. We report the results of IMvigor130, a phase 3 trial that compared atezolizumab with or without platinum-based chemotherapy versus placebo plus platinum-based chemotherapy in first-line metastatic urothelial carcinoma. METHODS: In this multicentre, phase 3, randomised trial, untreated patients aged 18 years or older with locally advanced or metastatic urothelial carcinoma, from 221 sites in 35 countries, were randomly assigned to receive atezolizumab plus platinum-based chemotherapy (group A), atezolizumab monotherapy (group B), or placebo plus platinum-based chemotherapy (group C). Patients received 21-day cycles of gemcitabine (1000 mg/m2 body surface area, administered intravenously on days 1 and 8 of each cycle), plus either carboplatin (area under the curve of 4·5 mg/mL per min administered intravenously) or cisplatin (70 mg/m2 body surface area administered intravenously) on day 1 of each cycle with either atezolizumab (1200 mg administered intravenously on day 1 of each cycle) or placebo. Group B patients received 1200 mg atezolizumab, administered intravenously on day 1 of each 21-day cycle. The co-primary efficacy endpoints for the intention-to-treat population were investigator-assessed Response Evaluation Criteria in Solid Tumours 1.1 progression-free survival and overall survival (group A vs group C) and overall survival (group B vs group C), which was to be formally tested only if overall survival was positive for group A versus group C. The trial is registered with ClinicalTrials.gov, NCT02807636. FINDINGS: Between July 15, 2016, and July 20, 2018, we enrolled 1213 patients. 451 (37%) were randomly assigned to group A, 362 (30%) to group B, and 400 (33%) to group C. Median follow-up for survival was 11·8 months (IQR 6·1-17·2) for all patients. At the time of final progression-free survival analysis and interim overall survival analysis (May 31, 2019), median progression-free survival in the intention-to-treat population was 8·2 months (95% CI 6·5-8·3) in group A and 6·3 months (6·2-7·0) in group C (stratified hazard ratio [HR] 0·82, 95% CI 0·70-0·96; one-sided p=0·007). Median overall survival was 16·0 months (13·9-18·9) in group A and 13·4 months (12·0-15·2) in group C (0·83, 0·69-1·00; one-sided p=0·027). Median overall survival was 15·7 months (13·1-17·8) for group B and 13·1 months (11·7-15·1) for group C (1·02, 0·83-1·24). Adverse events that led to withdrawal of any agent occurred in 156 (34%) patients in group A, 22 (6%) patients in group B, and 132 (34%) patients in group C. 50 (11%) patients in group A, 21 (6%) patients in group B, and 27 (7%) patients in group C had adverse events that led to discontinuation of atezolizumab or placebo. INTERPRETATION: Addition of atezolizumab to platinum-based chemotherapy as first-line treatment prolonged progression-free survival in patients with metastatic urothelial carcinoma. The safety profile of the combination was consistent with that observed with the individual agents. These results support the use of atezolizumab plus platinum-based chemotherapy as a potential first-line treatment option for metastatic urothelial carcinoma. FUNDING: F Hoffmann-La Roche and Genentech.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Transitional Cell/drug therapy , Cisplatin/administration & dosage , Urologic Neoplasms/drug therapy , Administration, Intravenous , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/adverse effects , Carcinoma, Transitional Cell/mortality , Cisplatin/adverse effects , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Progression-Free Survival , Proportional Hazards Models , Urologic Neoplasms/mortality
2.
Cancer Chemother Pharmacol ; 84(6): 1257-1267, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31542806

ABSTRACT

PURPOSE: To determine the exposure-response (ER) relationships between atezolizumab exposure and efficacy or safety in patients with advanced non-small cell lung cancer (NSCLC) or urothelial carcinoma (UC) and to identify alternative dosing regimens. METHODS: ER analyses were conducted using pooled NSCLC and UC data from phase 1 and 3 studies (PCD4989g, OAK, IMvigor211; ClinicalTrials.gov IDs, NCT01375842, NCT02008227, and NCT02302807, respectively). Objective response rate, overall survival, and adverse events were evaluated vs pharmacokinetic (PK) metrics. Population PK-simulated exposures for regimens of 840 mg every 2 weeks (q2w) and 1680 mg every 4 weeks (q4w) were compared with the approved regimen of 1200 mg every 3 weeks (q3w) and the maximum assessed dose (MAD; 20 mg/kg q3w). Phase 3 IMpassion130 (NCT02425891) data were used to validate the PK simulations for 840 mg q2w. Observed safety data were evaluated by exposure and body weight subgroups. RESULTS: No significant ER relationships were observed for safety or efficacy. Predicted exposures for 840 mg q2w and 1680 mg q4w were comparable to 1200 mg q3w and the MAD and consistent with observed PK data from IMpassion130. Observed safety was similar between patients with a Cmax above and below the predicted Cmax for 1680 mg q4w and between patients in the lowest and upper 3 body weight quartiles. CONCLUSION: Atezolizumab regimens of 840 mg q2w and 1680 mg q4w are expected to have comparable efficacy and safety as the approved regimen of 1200 mg q3w, supporting their interchangeable use and offering patients greater flexibility.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Transitional Cell/drug therapy , Lung Neoplasms/drug therapy , Models, Biological , Urologic Neoplasms/drug therapy , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Transitional Cell/mortality , Clinical Trials, Phase I as Topic , Clinical Trials, Phase III as Topic , Computer Simulation , Datasets as Topic , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Lung Neoplasms/mortality , Male , Monte Carlo Method , Randomized Controlled Trials as Topic , Survival Analysis , Treatment Outcome , Urologic Neoplasms/mortality
3.
J Comp Eff Res ; 8(14): 1173-1185, 2019 10.
Article in English | MEDLINE | ID: mdl-31394922

ABSTRACT

Aim: This observational study evaluated the effectiveness of nab-paclitaxel versus paclitaxel monotherapy as first-line (1L) treatment for metastatic triple-negative breast cancer (mTNBC). Materials & methods: 200 patients from the US Flatiron Health electronic health record-derived database (mTNBC diagnosis, January 2011-October 2016) who received 1L nab-paclitaxel (n = 105) or paclitaxel (n = 95) monotherapy were included. Overall survival and time to next treatment were evaluated. Results: The adjusted overall survival hazard ratio was 0.98 (95% CI: 0.67-1.44), indicating a similar risk of death between groups. Adjusted time to next treatment hazard ratio was 0.89 (95% confidence interval: 0.62-1.29). Conclusion: Nab-paclitaxel and paclitaxel monotherapy showed similar efficacy, suggesting their interchangeability as 1L treatments for mTNBC.


Subject(s)
Paclitaxel/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Aged , Albumins/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Female , Humans , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Survival Analysis , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
4.
Acc Chem Res ; 40(5): 335-42, 2007 May.
Article in English | MEDLINE | ID: mdl-17474708

ABSTRACT

Polycationic organic nanoparticles are shown to disrupt model biological membranes and living cell membranes at nanomolar concentrations. The degree of disruption is shown to be related to nanoparticle size and charge, as well as to the phase-fluid, liquid crystalline, or gel-of the biological membrane. Disruption events on model membranes have been directly imaged using scanning probe microscopy, whereas disruption events on living cells have been analyzed using cytosolic enzyme leakage assays, dye diffusion assays, and fluorescence microscopy.


Subject(s)
Cell Membrane/metabolism , Lipid Bilayers/chemistry , Nanoparticles/chemistry , Nanotechnology , Cell Membrane/chemistry , Cell Membrane Permeability , Cytosol/chemistry , Cytosol/metabolism , Diffusion , Dimyristoylphosphatidylcholine/chemistry , Dimyristoylphosphatidylcholine/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Lipid Bilayers/metabolism , Microscopy, Atomic Force , Microscopy, Fluorescence , Models, Biological , Models, Molecular , Particle Size
5.
Soft Matter ; 2(9): 751-759, 2006 Aug 16.
Article in English | MEDLINE | ID: mdl-32680215

ABSTRACT

A review dedicated to block copolymer self assembly. We discuss general progress in physicochemical interpretations and provide insight to recent developments in (hybrid) materials.

6.
Langmuir ; 21(23): 10348-54, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16262291

ABSTRACT

Polycationic polymers are used extensively in biology to disrupt cell membranes and thus enhance the transport of materials into the cell. The highly polydisperse nature of many of these materials makes obtaining a mechanistic understanding of the disruption processes difficult. To design an effective mechanistic study, a monodisperse class of polycationic polymers, poly(amidoamine) (PAMAM) dendrimers, has been studied in the context of supported dimyristoylphosphatidylcholine (DMPC) lipid bilayers using atomic force microscopy (AFM). Aqueous solutions of amine-terminated generation 7 (G7) PAMAM dendrimers caused the formation of 15-40-nm-diameter holes in lipid bilayers. This effect was significantly reduced for smaller G5 dendrimers. For G3, no hole formation was observed. In addition to dendrimer size, surface chemistry had a strong influence on dendrimer-lipid bilayer interactions. In particular, acetamide-terminated G5 did not cause hole formation in bilayers. In all instances, the edges of bilayer defects proved to be points of highest dendrimer activity. A proposed mechanism for the removal of lipids by dendrimers involves the formation of dendrimer-filled lipid vesicles. By considering the thermodynamics, interaction free energy, and geometry of these self-assembled vesicles, a model that explains the influence of polymer particle size and surface chemistry on the interactions with lipid membranes was developed. These results are of general significance for understanding the physical and chemical properties of polycationic polymer interactions with membranes that lead to the transport of materials across cell membranes.


Subject(s)
Lipid Bilayers , Polymers , Cations , Microscopy, Atomic Force
7.
Langmuir ; 21(20): 9280-6, 2005 Sep 27.
Article in English | MEDLINE | ID: mdl-16171363

ABSTRACT

Apoptosis is defined by a distinct set of morphological changes observed during cell death including loss of focal adhesions, the formation of cell membrane buds or blebs, and a decrease in total cell volume. Recent studies suggest that these dramatic morphological changes, particularly apoptotic volume decrease (AVD), are an early prerequisite to apoptosis and precede key biochemical time-points. Here we use atomic force microscopy to observe early stage AVD of KB cells undergoing staurosporine-induced apoptosis. After a 3-h exposure to 1 microM staurosporine, a 32% decrease in total cell height and a 50% loss of total cell volume is observed accompanied by only a 15% change in cell diameter. The observed AVD precedes key biochemical hallmarks of apoptosis such as loss of mitochondrial membrane potential, phosphatidyl serine translocation, nuclear fragmentation, and measurable caspase-3 activity. This suggests that morphological volume changes occur very early in the induction of apoptosis.


Subject(s)
Apoptosis/physiology , Cell Membrane/ultrastructure , Cell Nucleus/ultrastructure , KB Cells/ultrastructure , Microscopy, Atomic Force/methods , Mitochondria/ultrastructure , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Size , Humans , KB Cells/metabolism , Mitochondria/metabolism , Phosphatidylserines/metabolism , Time Factors
8.
Langmuir ; 21(19): 8588-90, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-16142931

ABSTRACT

Polycationic polymers are known to disrupt lipid bilayers. In this letter, we report the dependence of this disruption on the lipid structural phase. DMPC bilayers are exposed to two polycationic polymeric nanoparticles, PAMAM dendrimers and MSI-78. We find that regions of the bilayer that are in the gel phase are unaffected by the presence of polymers, whereas the liquid phase is disrupted.


Subject(s)
Dimyristoylphosphatidylcholine/chemistry , Lipid Bilayers/chemistry , Nanoparticles/chemistry , Polyamines/chemical synthesis , Polymers/chemical synthesis , Antimicrobial Cationic Peptides/chemistry , Dendrimers , Particle Size , Polyamines/chemistry , Polyelectrolytes , Polymers/chemistry , Surface Properties
9.
Biophys J ; 89(6): 4043-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16183881

ABSTRACT

The interaction of an antimicrobial peptide, MSI-78, with phospholipid bilayers has been investigated using atomic force microscopy, circular dichroism, and nuclear magnetic resonance (NMR). Binding of amphipathic peptide helices with their helical axis parallel to the membrane surface leads to membrane thinning. Atomic force microscopy of supported 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) bilayers in the presence of MSI-78 provides images of the membrane thinning process at a high spatial resolution. This data reveals that the membrane thickness is not reduced uniformly over the entire bilayer area. Instead, peptide binding leads to the formation of distinct domains where the bilayer thickness is reduced by 1.1 +/- 0.2 nm. The data is interpreted using a previously published geometric model for the structure of the peptide-lipid domains. In this model, the peptides reside at the hydrophilic-hydrophobic boundary in the lipid headgroup region, which leads to an increased distance between lipid headgroups. This picture is consistent with concentration-dependent 31P and 2H NMR spectra of MSI-78 in mechanically aligned DMPC bilayers. Furthermore, 2H NMR experiments on DMPC-d54 multilamellar vesicles indicate that the acyl chains of DMPC are highly disordered in the presence of the peptide as is to be expected for the proposed structure of the peptide-lipid assembly.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Lipid Bilayers/chemistry , Liposomes/chemistry , Membrane Fluidity , Membrane Microdomains/chemistry , Membrane Microdomains/ultrastructure , Microscopy, Atomic Force/methods , Molecular Conformation , Protein Binding
10.
Chem Phys Lipids ; 132(1): 3-14, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15530443

ABSTRACT

Atomic force microscopy (AFM) is employed to observe the effect of poly(amidoamine) (PAMAM) dendrimers on 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) lipid bilayers. Aqueous solutions of generation 7 PAMAM dendrimers cause the formation of holes 15-40 nm in diameter in previously intact bilayers. This effect is observed for two different branch end-groups--amine and carboxyl. In contrast, carboxyl-terminated core-shell tectodendrimer clusters do not create holes in the lipid membrane but instead show a strong affinity to adsorb to the edges of existing bilayer defects. A possible mechanism for the formation of holes in the lipid bilayer is proposed. The dendrimers remove lipid molecules from the substrate and form aggregates consisting of a dendrimer surrounded by lipid molecules. Dynamic light scattering (DLS) measurements as well as 31P NMR data support this explanation. The fact that tectodendrimers behave differently suggests that their cluster-like architecture plays an important role in their interaction with the lipid bilayer.


Subject(s)
Dimyristoylphosphatidylcholine/chemistry , Lipid Bilayers/chemistry , Liposomes/chemistry , Membrane Fluidity , Polyamines/chemistry , Macromolecular Substances , Magnetic Resonance Spectroscopy , Membranes, Artificial , Microscopy, Atomic Force , Particle Size , Surface Properties
11.
Bioconjug Chem ; 15(4): 774-82, 2004.
Article in English | MEDLINE | ID: mdl-15264864

ABSTRACT

We have investigated poly(amidoamine) (PAMAM) dendrimer interactions with supported 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) lipid bilayers and KB and Rat2 cell membranes using atomic force microscopy (AFM), enzyme assays, flow cell cytometry, and fluorescence microscopy. Amine-terminated generation 7 (G7) PAMAM dendrimers (10-100 nM) were observed to form holes of 15-40 nm in diameter in aqueous, supported lipid bilayers. G5 amine-terminated dendrimers did not initiate hole formation but expanded holes at existing defects. Acetamide-terminated G5 PAMAM dendrimers did not cause hole formation in this concentration range. The interactions between PAMAM dendrimers and cell membranes were studied in vitro using KB and Rat 2 cell lines. Neither G5 amine- nor acetamide-terminated PAMAM dendrimers were cytotoxic up to a 500 nM concentration. However, the dose dependent release of the cytoplasmic proteins lactate dehydrogenase (LDH) and luciferase (Luc) indicated that the presence of the amine-terminated G5 PAMAM dendrimer decreased the integrity of the cell membrane. In contrast, the presence of acetamide-terminated G5 PAMAM dendrimer had little effect on membrane integrity up to a 500 nM concentration. The induction of permeability caused by the amine-terminated dendrimers was not permanent, and leaking of cytosolic enzymes returned to normal levels upon removal of the dendrimers. The mechanism of how PAMAM dendrimers altered cells was investigated using fluorescence microscopy, LDH and Luc assays, and flow cytometry. This study revealed that (1) a hole formation mechanism is consistent with the observations of dendrimer internalization, (2) cytosolic proteins can diffuse out of the cell via these holes, and (3) dye molecules can be detected diffusing into the cell or out of the cell through the same membrane holes. Diffusion of dendrimers through holes is sufficient to explain the uptake of G5 amine-terminated PAMAM dendrimers into cells and is consistent with the lack of uptake of G5 acetamide-terminated PAMAM dendrimers.


Subject(s)
Amides/chemistry , Amides/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Polyamines/chemistry , Polyamines/metabolism , Polymers/chemistry , Polymers/metabolism , Acetamides/chemistry , Acetamides/metabolism , Amines/chemistry , Amines/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane Permeability , Coloring Agents/chemistry , Coloring Agents/metabolism , Diffusion , Dimyristoylphosphatidylcholine/chemistry , Dimyristoylphosphatidylcholine/metabolism , Drug Delivery Systems , Flow Cytometry , Luciferases/metabolism , Microscopy, Atomic Force , Molecular Structure , Rats
SELECTION OF CITATIONS
SEARCH DETAIL