Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
JTO Clin Res Rep ; 3(2): 100275, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35146462

ABSTRACT

INTRODUCTION: Programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors are standard-of-care treatment for metastatic NSCLC (mNSCLC). Intolerance to treatment/disease progression warrants additional lines of therapy. Real-world treatment patterns and efficacy outcomes after PD-1/PD-L1 use are insufficiently characterized to inform treatment decisions. METHODS: Electronic health records of adults with stage IV NSCLC initiating PD-1/PD-L1 inhibitors as first-line monotherapy (cohort 1), first-line combination therapy (cohort 2), or second-line monotherapy (cohort 3) who received a subsequent line of therapy (i.e., index therapy) in the Flatiron NSCLC Core Registry Dataset were identified. Patient characteristics, types of index treatments/therapies, and associated index treatment outcomes were extracted. RESULTS: A total of 1061 patients with mNSCLC were included in this analysis. In cohort 1 (n = 242), median real-world overall survival (mrwOS) with index therapies for the overall population was 9.18 months (95% confidence interval: 7.54-12.13); platinum-based chemotherapy was the most common index therapy (39.3%) with mrwOS of 12.52 months (8.39-not applicable). In cohort 2 (n = 145), mrwOS for the overall population was 6.43 months (5.34-7.61); vascular endothelial growth factor inhibitor plus chemotherapy was the most common index therapy (32.4%) with mrwOS of 5.97 months (4.95-7.34). In cohort 3 (n = 647), mrwOS for the overall population was 7.21 months (6.39-7.80); single-agent chemotherapy was the most common index therapy (45.4%) with mrwOS of 6.59 months (5.64-7.61). CONCLUSIONS: Real-world treatment patterns and survival outcomes of index therapies in mNSCLC after PD-1/PD-L1 use are variable. These analyses provide insights to optimize post-PD-1/PD-L1 treatments and inform standards of care.

2.
Front Oncol ; 8: 253, 2018.
Article in English | MEDLINE | ID: mdl-30087850

ABSTRACT

INTRODUCTION: The phase II ABOUND.PS2 study (NCT02289456) assessed safety/tolerability of a first-line modified nab-paclitaxel/carboplatin regimen for patients with advanced non-small cell lung cancer (NSCLC) and Eastern Cooperative Oncology Group (ECOG) performance status (PS) 2. METHODS: Chemotherapy-naive patients with stage IIIB/IV NSCLC and ECOG PS 2 received four cycles of nab-paclitaxel 100 mg/m2 days 1 and 8 plus carboplatin area under the curve 5 day 1 q3w (induction). Patients without progression received nab-paclitaxel monotherapy (100 mg/m2 days 1 and 8 q3w) until progression/unacceptable toxicity. Primary endpoint: percentage of patients discontinuing induction due to treatment-emergent adverse events (TEAEs). RESULTS: 11/40 treated patients (27.5%; 95% CI, 14.60-43.89) discontinued chemotherapy induction due to TEAEs; 16/40 (40.0%) continued nab-paclitaxel monotherapy. Median progression-free and overall survival were 4.4 (95% CI, 2.99-7.00) and 7.7 (95% CI, 4.93-13.17) months. Grade 3/4 TEAEs during induction included neutropenia (22.5%), anemia (17.5%), thrombocytopenia (5.0%), and peripheral neuropathy (2.5%). CONCLUSION: This nab-paclitaxel-based regimen was tolerable in patients with advanced NSCLC and ECOG PS 2, with efficacy comparable to historical chemotherapy data.

3.
Front Oncol ; 8: 262, 2018.
Article in English | MEDLINE | ID: mdl-30087851

ABSTRACT

The phase 4 ABOUND.70+ trial assessed the safety and efficacy of nab-paclitaxel/carboplatin continuously or with a 1-week break between cycles in elderly patients with advanced non-small cell lung cancer (NSCLC). Patients ≥70 years with locally advanced/metastatic NSCLC were randomized 1:1 to first-line nab-paclitaxel days 1, 8, 15 plus carboplatin day 1 of a 21-day cycle (21d) or the same nab-paclitaxel/carboplatin regimen with a 1-week break between cycles (21d + break; 28d). The primary endpoint was the percentage of patients with grade ≥ 2 peripheral neuropathy (PN) or grade ≥ 3 myelosuppression. Other key endpoints included progression-free survival (PFS), overall survival (OS), and overall response rate (ORR). A total of 143 patients were randomized (71 to 21d, 72 to 21d + break). The percentage of patients with grade ≥ 2 PN or grade ≥ 3 myelosuppression was similar between the 21d and 21d + break arms (76.5 and 77.1%; P = 0.9258). Treatment exposure was lower in the 21d arm compared with the 21d + break arm. Median OS was 15.2 and 16.2 months [hazard ratio (HR) 0.72, 95% CI 0.44-1.19; P = 0.1966], median PFS was 3.6 and 7.0 months (HR 0.48, 95% CI 0.30-0.76; P < 0.0019), and ORR was 23.9 and 40.3% (risk ratio 1.68, 95% CI 1.02-2.78; P = 0.0376) in the 21d and 21d + break arms, respectively. In summary, the 1-week break between treatment cycles significantly improved PFS and ORR but did not significantly reduce the percentage of grade ≥ 2 PN or grade ≥ 3 myelosuppression. Overall, the findings support the results of prior subset analyses on the safety and efficacy of first-line nab-paclitaxel/carboplatin in elderly patients with advanced NSCLC.

4.
Invest New Drugs ; 32(5): 1017-27, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24890858

ABSTRACT

PURPOSE: To evaluate the efficacy and tolerability of bortezomib in combination with doxorubicin in patients with advanced hepatocellular carcinoma, and to correlate pharmacodynamic markers of proteasome inhibition with response and survival. EXPERIMENTAL DESIGN: This phase II, open-label, multicenter study examined the efficacy of bortezomib (1.3 mg/m(2) IV on d1, 4, 8, 11) and doxorubicin (15 mg/m(2) IV on d1, 8) in 21-day cycles. The primary endpoint was objective response rate. RESULTS: Best responses in 38 treated patients were 1 partial response (2.6 %), 10 (26.3 %) stable disease, and 17 (44.7 %) progressive disease; 10 patients were unevaluable. Median PFS was 2.2 months. Median OS was 6.1 months. The most common grade 3 to 4 toxicities were hypertension, glucose intolerance, ascites, ALT elevation, hyperglycemia and thrombosis/embolism. Worse PFS was seen in patients with elevated IL-6, IL-8, MIP-1α and EMSA for NF-κB at the start of treatment. Worse OS was seen in patients with elevated IL-8 and VEGF at the start of treatment. Patients had improved OS if a change in the natural log of serum MIP-1α/CCL3 was seen after treatment. RANTES/CCL5 levels decreased significantly with treatment. CONCLUSIONS: The combination of doxorubicin and bortezomib was well-tolerated in patients with hepatocellular carcinoma, but the primary endpoint was not met. Exploratory analyses of markers of proteasome inhibition suggest a possible prognostic and predictive role and should be explored further in tumor types for which bortezomib is efficacious.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Boronic Acids/pharmacology , Bortezomib , Carcinoma, Hepatocellular/blood , Chemokine CCL5/blood , Cytokines/blood , Disease-Free Survival , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Female , Humans , Liver Neoplasms/blood , Male , Middle Aged , Pyrazines/administration & dosage , Pyrazines/adverse effects , Pyrazines/pharmacology , Treatment Outcome , Vascular Endothelial Growth Factor A/blood , Young Adult
5.
EMBO Mol Med ; 5(1): 149-66, 2013 01.
Article in English | MEDLINE | ID: mdl-23180582

ABSTRACT

Oncogene-induced senescence can provide a protective mechanism against tumour progression. However, production of cytokines and growth factors by senescent cells may contribute to tumour development. Thus, it is unclear whether induction of senescence represents a viable therapeutic approach. Here, using a mouse model with orthotopic implantation of metastatic melanoma tumours taken from 19 patients, we observed that targeting aurora kinases with MLN8054/MLN8237 impaired mitosis, induced senescence and markedly blocked proliferation in patient tumour implants. Importantly, when a subset of tumour-bearing mice were monitored for tumour progression after pausing MLN8054 treatment, 50% of the tumours did not progress over a 12-month period. Mechanistic analyses revealed that inhibition of aurora kinases induced polyploidy and the ATM/Chk2 DNA damage response, which mediated senescence and a NF-κB-related, senescence-associated secretory phenotype (SASP). Blockade of IKKß/NF-κB led to reversal of MLN8237-induced senescence and SASP. Results demonstrate that removal of senescent tumour cells by infiltrating myeloid cells is crucial for inhibition of tumour re-growth. Altogether, these data demonstrate that induction of senescence, coupled with immune surveillance, can limit melanoma growth.


Subject(s)
Azepines/pharmacology , Benzazepines/pharmacology , Melanoma, Experimental/drug therapy , NF-kappa B/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins , Aurora Kinases , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Checkpoint Kinase 2 , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Nude , Polyploidy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 18(8): 2184-98, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22351689

ABSTRACT

PURPOSE: The purpose of this preclinical study was to determine the effectiveness of RAF265, a multikinase inhibitor, for treatment of human metastatic melanoma and to characterize traits associated with drug response. EXPERIMENTAL DESIGN: Advanced metastatic melanoma tumors from 34 patients were orthotopically implanted to nude mice. Tumors that grew in mice (17 of 34) were evaluated for response to RAF265 (40 mg/kg, every day) over 30 days. The relation between patient characteristics, gene mutation profile, global gene expression profile, and RAF265 effects on tumor growth, mitogen-activated protein/extracellular signal-regulated kinase (MEK)/extracellular signal-regulated kinase (ERK) phosphorylation, proliferation, and apoptosis markers was evaluated. RESULTS: Nine of the 17 tumors that successfully implanted (53%) were mutant BRAF (BRAF(V600E/K)), whereas eight of 17 (47%) tumors were BRAF wild type (BRAF(WT)). Tumor implants from 7 of 17 patients (41%) responded to RAF265 treatment with more than 50% reduction in tumor growth. Five of the 7 (71%) responders were BRAF(WT), of which 1 carried c-KIT(L576P) and another N-RAS(Q61R) mutation, while only 2 (29%) of the responding tumors were BRAF(V600E/K). Gene expression microarray data from nonimplanted tumors revealed that responders exhibited enriched expression of genes involved in cell growth, proliferation, development, cell signaling, gene expression, and cancer pathways. Although response to RAF265 did not correlate with pERK1/2 reduction, RAF265 responders did exhibit reduced pMEK1, reduced proliferation based upon reduced Ki-67, cyclin D1 and polo-like kinase1 levels, and induction of the apoptosis mediator BCL2-like 11. CONCLUSIONS: Orthotopic implants of patient tumors in mice may predict prognosis and treatment response for melanoma patients. A subpopulation of human melanoma tumors responds to RAF265 and can be characterized by gene mutation and gene expression profiles.


Subject(s)
Imidazoles/pharmacology , Imidazoles/therapeutic use , Melanoma/drug therapy , Pyridines/pharmacology , Pyridines/therapeutic use , Animals , Apoptosis/drug effects , Base Sequence , Cell Cycle Proteins/biosynthesis , Cell Proliferation/drug effects , Cyclin D1/biosynthesis , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Gene Expression Profiling , Humans , Ki-67 Antigen/biosynthesis , MAP Kinase Signaling System/drug effects , Male , Melanoma/metabolism , Melanoma/pathology , Melanoma/secondary , Mice , Mice, Nude , Mutation , Oligonucleotide Array Sequence Analysis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Analysis, DNA , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , bcl-X Protein/biosynthesis , Polo-Like Kinase 1
7.
Chem Commun (Camb) ; 46(30): 5557-9, 2010 Aug 14.
Article in English | MEDLINE | ID: mdl-20526488

ABSTRACT

A new intracellular mRNA imaging probe has been developed that incorporates thiol-terminated hairpin oligonucleotides covalently bound to the surface of citrate-capped gold nanoparticles. The hairpin DNA-coated gold nanoparticles (hAuNPs) positively identifies tyrosinase mRNA in cultured melanoma cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Inverted Repeat Sequences , Melanoma/diagnosis , Monophenol Monooxygenase/genetics , Nanoparticles , Oligonucleotides , RNA, Messenger/analysis , Cell Line, Tumor , Gold/chemistry , Humans , Melanoma/genetics , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Nanoparticles/chemistry , Oligonucleotides/chemistry , RNA, Messenger/genetics , Sulfhydryl Compounds/chemistry
8.
Clin Cancer Res ; 16(1): 348-57, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028756

ABSTRACT

PURPOSE: Preclinical studies show that bortezomib, a proteasome inhibitor, blocks NF-kappaB activation and, combined with temozolomide, enhances activity against human melanoma xenografts and modulates other critical tumor targets. We initiated a phase I trial of temozolomide plus bortezomib in advanced melanoma. Objectives included defining a maximum tolerated dose for the combination, characterizing biomarker changes reflecting inhibition of both proteasome and NF-kappaB activity in blood (if possible tumor), and characterizing antitumor activity. EXPERIMENTAL DESIGN: Cohorts were enrolled onto escalating dose levels of temozolomide (50-75 mg/m(2)) daily, orally, for 6 of 9 weeks and bortezomib (0.75-1.5 mg/m(2)) by i.v. push on days 1, 4, 8, and 11 every 21 days. Peripheral blood mononuclear cells were assayed at specified time points for proteasome inhibition and NF-kappaB biomarker activity. RESULTS: Bortezomib (1.3 mg/m(2)) and temozolomide (75 mg/m(2)) proved to be the maximum tolerated dose. Dose-limiting toxicities included neurotoxicity, fatigue, diarrhea, and rash. Nineteen melanoma patients were enrolled onto four dose levels. This melanoma population (17 M1c, 10 elevated lactate dehydrogenase, 12 performance status 1-2) showed only one partial response (8 months) and three with stable disease >or=4 months. A significant reduction in proteasome-specific activity was observed 1 hour after infusion at all bortezomib doses. Changes in NF-kappaB electrophoretic mobility shift assay and circulating chemokines in blood failed to correlate with the schedule/dose of bortezomib, inhibition of proteasome activity, or clinical outcome. CONCLUSIONS: We have defined phase II doses for this schedule of temozolomide with bortezomib. Although proteasome activity was inhibited for a limited time in peripheral blood mononuclear cells, we were unable to show consistent effects on NF-kappaB activation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/administration & dosage , Dacarbazine/analogs & derivatives , Melanoma/drug therapy , Pyrazines/administration & dosage , Skin Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/adverse effects , Bortezomib , Chemokines/blood , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Drug Administration Schedule , Female , Humans , Male , Melanoma/blood , Middle Aged , NF-kappa B/antagonists & inhibitors , NF-kappa B/blood , Proteasome Endopeptidase Complex/blood , Proteasome Inhibitors , Pyrazines/adverse effects , Skin Neoplasms/blood , Temozolomide , Treatment Outcome
9.
Clin Cancer Res ; 12(3 Pt 1): 950-60, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16467110

ABSTRACT

PURPOSE: Constitutive activation of inhibitor of kappaB kinase (IKK) confers melanoma resistance to apoptosis and chemotherapy. Whether IKK is able to serve as a therapeutic target in melanoma is unknown. We explored the possibility of exploiting IKK as a therapeutic target in melanoma by using BMS-345541, a novel compound with a highly selective IKKbeta inhibitory activity, to trigger melanoma cell apoptosis. EXPERIMENTAL DESIGN: Three human melanoma cell lines (SK-MEL-5, Hs 294T, and A375), all of which have high constitutive IKK activities, served as in vitro and in vivo melanoma models for treatment with BMS-345541. Two known antitumor drugs (temozolomide and bortezomib) were used as parallel controls for evaluation of the therapeutic efficiency and toxicity of BMS-345541. The effects of BMS-345541 on nuclear factor kappaB (NF-kappaB) signaling and on the apoptosis machinery were investigated. RESULTS: Inhibition of constitutive IKK activity by BMS-345541 resulted in the reduction of NF-kappaB activity, CXCL1 chemokine secretion by cultured melanoma cells and melanoma cell survival in vitro and in vivo. The effect of BMS-345541 on tumor cell growth was through mitochondria-mediated apoptosis, based on the release of apoptosis-inducing factor, dissipation of mitochondrial membrane potential, and reduced ratio of B cell lymphoma gene-2 (Bcl-2)/Bcl-associated X protein (Bax) in mitochondria. The BMS-345541 execution of apoptosis was apoptosis-inducing factor-dependent, but largely caspase-independent. CONCLUSION: BMS-345541 down-regulation of IKK activity results in mitochondria-mediated apoptosis of tumor cells because the programmed cell death machinery in melanoma cells is highly regulated by NF-kappaB signaling. Therefore, IKK may serve as a potential target for melanoma therapy.


Subject(s)
Apoptosis/drug effects , I-kappa B Kinase/antagonists & inhibitors , Imidazoles/pharmacology , Melanoma/enzymology , Melanoma/metabolism , Mitochondria/metabolism , NF-kappa B/metabolism , Quinoxalines/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Imidazoles/administration & dosage , Imidazoles/therapeutic use , In Vitro Techniques , Melanoma/drug therapy , Mice , Mitochondria/drug effects , NF-kappa B/drug effects , Quinoxalines/administration & dosage , Quinoxalines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , Structure-Activity Relationship , Time Factors , Tumor Cells, Cultured
10.
Cancer Metastasis Rev ; 24(2): 301-13, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15986139

ABSTRACT

Nuclear Factor-kappa B (NF-kappa B) is an inducible transcription factor that regulates the expression of many genes involved in the immune response. Recently, NF-kappa B activity has been shown to be upregulated in many cancers, including melanoma. Data indicate that the enhanced activation of NF-kappa B may be due to deregulations in upstream signaling pathways such as Ras/Raf, PI3K/Akt, and NIK. Multiple studies have shown that NF-kappa B is involved in the regulation of apoptosis, angiogenesis, and tumor cell invasion, all of which indicate the important role of NF-kappa B in tumorigenesis. Thus, understanding the molecular mechanism of melanoma progression will aid in designing new therapeutic approaches for melanoma. In this review, the association between NF-kappa B and melanoma tumorigenesis are discussed. Additionally, the potential of emerging selective NF-kappa B inhibitors for the treatment of melanoma is reviewed.


Subject(s)
Cell Transformation, Neoplastic , Melanoma/physiopathology , NF-kappa B/physiology , Skin Neoplasms/physiopathology , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Melanoma/genetics , NF-kappa B/biosynthesis , NF-kappa B/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Up-Regulation
11.
Cancer Res ; 64(14): 4912-8, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15256463

ABSTRACT

Melanoma poses a great challenge to patients, oncologists, and biologists because of its nearly universal resistance to chemotherapy. Many studies have shown that nuclear factor kappaB is constitutively activated in melanoma, thereby promoting the proliferation of melanoma cells by inhibiting the apoptotic responses to chemotherapy. Nuclear factor kappaB activity is regulated by phosphorylation and subsequent degradation of inhibitor of nuclear factor kappaB by the ubiquitin-proteasome pathway. In this study, we show that the novel proteasome inhibitor, bortezomib, inhibited the growth of melanoma cells in vitro at a concentration range of 0.1-10 nM and in combination with the chemotherapeutic agent temozolomide, the inhibitory effect on melanoma cell growth was even more prominent. Data from a murine model showed reduced tumor growth when bortezomib was administered to human melanoma tumors. Strikingly, animals receiving bortezomib in combination with temozolomide achieved complete remission of palpable tumors after only 30 days of therapy, lasting >200 days. Our data indicate strongly that bortezomib in combination with chemotherapeutic agents should be studied additionally for the treatment of melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/pharmacology , Dacarbazine/analogs & derivatives , Melanoma/drug therapy , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Boronic Acids/administration & dosage , Bortezomib , Cell Division/drug effects , Cell Line, Tumor , Dacarbazine/administration & dosage , Drug Synergism , Female , Gene Expression/drug effects , Humans , Melanoma/blood supply , Melanoma/enzymology , Melanoma/pathology , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Neovascularization, Pathologic/drug therapy , Protease Inhibitors/administration & dosage , Pyrazines/administration & dosage , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL
...