Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Expert Opin Pharmacother ; 18(7): 701-716, 2017 May.
Article in English | MEDLINE | ID: mdl-28388240

ABSTRACT

INTRODUCTION: Tubulin inhibitors including taxanes and vinca alkaloids are important components of chemotherapy regimens used in advanced non-small cell lung cancer (NSCLC). Despite a treatment paradigm shift due to molecularly-targeted therapies and immunotherapy, a majority of patients will receive chemotherapy during their treatment course. Either used alone or in combination, tubulin inhibitors have demonstrated clinical benefits in different settings of lung cancer management. Areas covered: This review first discusses FDA-approved tubulin inhibitors for NSCLC, such as paclitaxel, docetaxel, vinorelbine, and nab-paclitaxel. The article then provides a summary of novel tubulin inhibitors, including cabazitaxel, eribulin, ixabepilone, patupilone, plinabulin, new colchicine analogues and others. It also discusses new tubulin inhibitor combinations with immunotherapy (PD-1/PD-L1 inhibitors) and molecularly-targeted therapies (e.g. anti-angiogenic agents, mTOR inhibitors, heat shock protein 90 inhibitors, MEK inhibitors, and anti-HER3 agents). Lastly, emerging data on potential resistance mechanisms and predictive biomarkers for tubulin inhibitors are explored. Expert opinion: Tubulin inhibitors will likely continue to play important roles in NSCLC management due to the advent of novel agents and combinations. Through further understanding of tumor biology, investigation of drug resistance, and development of predictive biomarkers, we will be better positioned to incorporate microtubule inhibition into patient specific treatment strategies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Tubulin Modulators/therapeutic use , Antineoplastic Agents/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans
2.
Ann Oncol ; 24(11): 2875-80, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24050956

ABSTRACT

BACKGROUND: Talactoferrin alfa is an oral dendritic cell (DC)-mediated immunotherapy (DCMI). We tested whether talactoferrin was superior to placebo in advanced non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: An FORTIS-M trial was an international, multicenter, randomized, double-blind comparison of talactoferrin (1.5 g p.o. BID) versus placebo BID, in patients with stage IIIB/IV NSCLC whose disease had failed two or more prior regimens. Treatment was administered for a maximum of five 14-week cycles. The primary efficacy end point was overall survival (OS); secondary end points included 6- and 12-month survival, progression-free survival (PFS), and disease control rate (DCR). RESULTS: Seven hundred and forty-two patients were randomly assigned (2:1) to talactoferrin (497) or placebo (245). The median OS in the intent-to-treat (ITT) population was 7.66 months in the placebo arm and 7.49 months in the talactoferrin arm [hazard ratio (HR), 1.04; 95% CI, 0.873-1.24; P = 0.6602]. The 6-month survival rates were 59.9% (95% CI, 53.4% to 65.8%) and 55.7% (95% CI, 51.1% to 59.9%), respectively. The 12-month survival rates were 32.2% (95% CI, 26.3% to 38.2%) and 30.9% (95% CI, 26.8% to 35%), respectively. The median PFS rates were 1.64 months and 1.68 months, respectively (HR, 0.99; 95% CI, 0.835-1.16; P = 0.8073). The DCRs were 38.4 and 37.6%, respectively [stratified odds ratio (OR), 0.96; 95% CI, 0.698-1.33; P = 0.8336]. The safety profiles were comparable between arms. CONCLUSIONS: There was no improvement in efficacy with talactoferrin alfa in patients with advanced NSCLC whose disease had failed two or more previous regimens.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Lactoferrin/administration & dosage , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Disease-Free Survival , Double-Blind Method , Drug Administration Schedule , Female , Humans , Immunotherapy , Kaplan-Meier Estimate , Lactoferrin/adverse effects , Male , Middle Aged , Neoplasm Staging , Placebos , Treatment Outcome
3.
Oncogene ; 28 Suppl 1: S38-45, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19680295

ABSTRACT

Despite advances in the management of non-small-cell lung cancer (NSCLC), including the introduction of targeted therapies such as epidermal growth factor receptor tyrosine kinase inhibitors, improvements in survival are marginal and the overall prognosis for patients remains poor. Tailoring therapy to the individual patient is a promising approach for selecting the most appropriate therapeutic regimens to maximize efficacy and minimize toxicity. The identification of predictive biomarkers that can guide treatment decisions is an important step for individualized therapy and in ultimately improving patient outcomes. Genomic and proteomic studies provide a means for the molecular profiling of tumor tissue from patients with NSCLC, and allow tailoring of therapy whereby the most appropriate treatment is administered to each individual patient. Although there are still significant challenges to implementing genomic and proteomic testing in clinical practice, the rapid development of newer technologies provides hope for overcoming these barriers.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Expression Profiling , Genomics , Humans , Pharmacogenetics , Prognosis , Proteomics , Treatment Outcome
4.
Cancer Chemother Pharmacol ; 47(6): 473-80, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11459199

ABSTRACT

The relationship between expression and function of the epidermal growth factor (EGF) family of receptors and chemosensitivity remains controversial. We studied the chemosensitivity to various anticancer agents of human cervical squamous carcinoma ME180 cells, and two resistant subclones, ME180/TNF and ME180/Pt, which also differ in their EGF receptor (EGFR) expression. Compared with ME180 cells, EGFR is overexpressed sixfold in ME180/TNF cells and is barely detectable in ME 180/Pt cells. Cell cycle analysis by flow cytometry and BrdU incorporation into DNA showed a correlation between EGFR expression and percentage of cells in S phase and active DNA replication (35% in high EGFR-expressing ME180/TNF cells, 19% in non-EGFR-expressing ME180/Pt cells and 23% in parental, intermediate-level EGFR-expressing ME 180 cells). By MTT assay and compared with parental, intermediate-level EGFR-expressing ME180 cells, high EGFR-expressing ME180/TNF cells had a three- to fourfold increased sensitivity to cisplatin, camptothecin (CPT), and topotecan, and low EGFR-expressing ME180/Pt cells had a five- to ninefold reduced sensitivity to the same agents. In contrast, the degree of cross-resistance with the topoisomerase II inhibitors doxorubicin and etoposide was minimal and the pattern of sensitivity to the anti-microtubulin agents vinblastine and paclitaxel was different, with a two- to fourfold decreased sensitivity in the high EGFR-expressing ME180/TNF cells and only a 1.5-fold decreased sensitivity in the low EGFR-expressing ME180/Pt cells. Neither alterations in intracellular CPT levels nor changes in topoisomerase I expression or activity, measured as ability to form DNA-protein complexes, were found to explain the differences in sensitivity to CPT among the three cell lines. Co-treatment with CP358774, a specific EGFR tyrosine kinase inhibitor, reduced the enhanced sensitivity of high EGFR-expressing ME180/TNF cells to the values observed in intermediate EGFR-expressing ME180 cells, but only reduced modestly the sensitivity of intermediate expressing ME180 cells. As a result, the resistance index of low EGFR-expressing ME180/Pt cells compared with intermediate EGFR-expressing ME180 cells was reduced only from five- to fourfold for cisplatin and from seven- to fourfold for CPT when ME180 cells were exposed to CP358774. CP358774 did not affect the sensitivity to either agent in low EGFR-expressing ME180/Pt cells. These results provide evidence that changes in EGFR expression or function may play a role in determining chemosensitivity to platinum and topoisomerase I poisons in some human tumor systems, and that the EGFR-related changes in chemosensitivity may vary depending on the level of EGFR expression and/or function.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Neoplasm Proteins/metabolism , Topotecan/pharmacology , Uterine Cervical Neoplasms/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Drug Resistance, Neoplasm , Female , Humans , Topoisomerase I Inhibitors , Tumor Cells, Cultured/drug effects , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
5.
Clin Cancer Res ; 7(6): 1683-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11410507

ABSTRACT

Abnormalities in the H-cadherin gene have been described in several human cancers. However, their biological significance remains undetermined. To investigate the role of H-cadherin in non-small cell lung cancer (NSCLC), a chimera H-cadherin-green fluorescent protein (GFP) expressed in Cos-7 cells was used to identify an anti-H- cadherin antibody, HCD-1. Western blot analysis was performed in six NSCLC cell lines and 35 pairs of primary NSCLC tumors and nonmalignant lung tissue obtained from surgical resections using HCD-1. Loss of H-cadherin expression was seen in five (83%) of the six NSCLC cell lines, whereas loss of E-cadherin was seen in three (50%) of the six. H-cadherin expression was lost in 15 (43%) of 35 NSCLC surgical tumor specimens, whereas E-cadherin expression was lost in 6 (17%) of 35. H-cadherin was expressed in all of the nonmalignant lung tissue from all of the surgical specimens. Fourteen of 35 tumors were heterotransplanted s.c. in nude mice. Tumorigenicity in nude mice was associated with both loss of H-cadherin expression (P = 0.03) and loss of E-cadherin expression (P = 0.05). Loss of H-cadherin was also associated with a more advanced local tumor growth, although the difference was not significant. The results indicate that loss of H-cadherin is frequent in human NSCLC and suggest that it facilitates the implantation and local growth of human NSCLC tumors.


Subject(s)
Cadherins/biosynthesis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Animals , Blotting, Western , COS Cells , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , RNA/metabolism , Recombinant Fusion Proteins/metabolism , Tissue Distribution , Tumor Cells, Cultured
6.
Clin Cancer Res ; 7(5): 1204-13, 2001 May.
Article in English | MEDLINE | ID: mdl-11350885

ABSTRACT

C225, a human-mouse chimerized monoclonal antibody directed against the epidermal growth factor receptor (EGFr), has a synergistic effect with cisplatin in xenograft models. To determine the tumor EGFr saturation dose with C225 and the fate of infused C225, we conducted a Phase Ib study with C225 in combination with cisplatin in patients with recurrent squamous cell carcinoma of the head and neck. Using tumor samples, we assessed tumor EGFr saturation by antibody using immunohistochemistry studies, the EGFr tyrosine kinase assay, and detection of the EGFr/C225 complex formation by immunoblot. Potential candidates were screened for EGFr expression in their tumors, and 12 patients who had high levels of EGFr expression and tumors easily accessible for repeated biopsies (pretherapy, 24 h after first C225 infusion, 24 h before third C225 infusion) were entered at three different dose levels of C225 with a fixed dose of cisplatin. The median value of tumor EGFr saturation increased to 95% at the higher dose levels. EGFr tyrosine kinase activity was significantly reduced after C225 infusion, and EGFr/C225 complexes were also detected at higher doses of C225. The loading dose of C225 at 400 mg/m(2) with a maintenance dose at 250 mg/m(2) achieved a high percentage of saturation of EGFr in tumor tissue, and these doses were recommended for Phases II or III clinical trials. Six (67%) of nine evaluable patients achieved major responses, including two (22%) complete responses. Mild to moderate degrees of allergic reaction and folliculitis-like skin reactions were demonstrated. We conclude that infused C225 binds and significantly saturates tumor EGFr, which may render a high degree of antitumor activity, and provides a novel mechanism for targeting cancer therapy for patients who have EGFr expression in their tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/metabolism , Head and Neck Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antigen-Antibody Complex/analysis , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cetuximab , Cisplatin/administration & dosage , Cisplatin/adverse effects , Female , Humans , Immunohistochemistry , Male , Middle Aged , Precipitin Tests , Treatment Outcome
7.
Mol Pharmacol ; 59(3): 593-603, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179455

ABSTRACT

Cell adhesion is important in the regulation of cell proliferation, migration, survival, and apoptosis. The major components of cell adhesion are the cadherin family of proteins, alpha-, beta- and gamma-catenins, and cytoskeletons. In addition, beta-catenin, when associated with adenomatous polyposis coli (APC) protein, an oncosuppressor, is implicated in the regulation of beta-catenin/APC-related signaling pathways. To examine the correlation between impairment of cell adhesion events and apoptosis, we used human non-small-cell lung cancer H460 and H520 cell lines as models to determine whether paclitaxel-induced apoptosis is associated with disruption of the components of cell adhesion and their functions. Paclitaxel treatment resulted in cells rounding up and losing contact with their neighboring cells, suggesting that the drug does indeed affect cell adhesion and related events. Western blot analysis revealed that paclitaxel caused a time- and concentration-dependent cleavage of beta-catenin, gamma-catenin, and APC protein, but not alpha-catenin or E-cadherin. These cleavages of beta-catenin and gamma-catenin were apoptosis-dependent, not mitosis-dependent. Paclitaxel treatment led to the proteolysis and activation of caspase-3 and -7, but not caspase-1. Furthermore, paclitaxel-induced apoptosis and cleavage of beta-catenin and gamma-catenin were inhibited by the pan-caspase inhibitor Z-VAD-FMK and partially inhibited by the caspase-3 inhibitor Z-DEVD-FMK but were not affected by the caspase-1 inhibitor AC-YVAD-CMK. Although the pan-caspase inhibitor blocked the cleavage of beta-catenin as well as DNA fragmentation, it did not affect paclitaxel-induced M-phase arrest and only partially prevented cell-growth inhibition. Biochemical studies revealed that cleaved beta-catenin was detected only in the Triton X-100 insoluble fraction, suggesting that it might localize in nuclear and/or membrane structures. Interestingly, the paclitaxel-induced beta-catenin fragment lost its ability to bind to E-cadherin, alpha-catenin, or APC protein and to serve as a substrate for tyrosine kinase. All our data demonstrate that the caspase-mediated cleavage of beta-catenin, gamma-catenin, and APC protein might contribute to paclitaxel-induced apoptosis.


Subject(s)
Apoptosis , Caspases/metabolism , Cell Adhesion/drug effects , Cytoskeletal Proteins/metabolism , Paclitaxel/pharmacology , Trans-Activators , Adenomatous Polyposis Coli Protein , Antineoplastic Agents, Phytogenic/pharmacology , Caspase 3 , Caspase 7 , Desmoplakins , Enzyme Activation , G2 Phase/drug effects , Humans , Mitosis/drug effects , Phosphorylation/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tumor Cells, Cultured , beta Catenin , gamma Catenin
8.
Cancer Chemother Pharmacol ; 46(5): 427-32, 2000.
Article in English | MEDLINE | ID: mdl-11127949

ABSTRACT

Annamycin is a highly lipophilic anthracycline with the ability to bypass the MDR-1 mechanism of cellular drug resistance. In this phase I study, annamycin entrapped in liposomes was administered by a 1- to 2-h intravenous infusion at 3-week intervals. Thirty-six patients with relapsed solid tumors were treated and 109 courses were administered at doses ranging from 3 to 240 mg/m2. The dose-limiting toxicity was thrombocytopenia. Five patients had a probable allergic reaction, requiring discontinuation of treatment in one. Treatment was well tolerated otherwise. No cardiac toxicity was seen on endomyocardial biopsy of four patients studied. There was limited gastrointestinal toxicity and no alopecia. No objective tumor responses were observed. Pharmacokinetic studies at 24, 120 and 240 mg/m2 showed a biexponential plasma concentration-versus-time profile. There was a linear relationship between the dose and the maximal plasma concentration with relatively constant plasma clearance values. The maximum tolerated dose (MTD) for liposomal annamycin defined in this study is 210 mg/m2. Because of a subsequent change in the formulation of the drug, future studies will use 190 mg/m2 as the MTD.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Blood Cell Count , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Carriers , Drug Resistance, Neoplasm , Female , Humans , Liposomes , Male , Middle Aged
9.
Cancer Biother Radiopharm ; 15(3): 253-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10941532

ABSTRACT

UNLABELLED: Cis-bis-neodecanoato-trans-R,R-1,2-diamminocyclohexane platinum (II) [NDDP] is a liposome-entrapped platinum compound currently, in phase II clinical trials, that has been shown to undergo intraliposomal activation. The objective of this study was to determine the feasibility of activating NDDP and using the induced radioactivity to monitor NDDP distribution and penetration. METHODS: Neutron activation analysis (NAA) was done on NDDP using the nuclear reactor at Texas A & M University, College Station, Texas. After a 3-hour irradiation, the NDDP samples were analyzed using an HPGE (high purity germanium) detector to determine the activation of the radioisotopic platinum. This was followed by HPLC-UV analysis to determine the stability of NDDP after exposure to the reactor's core. RESULTS: Platinum radioisotopes were produced along with potassium-40 and sodium-24. Irradiation did not result in any significant degradation of NDDP. CONCLUSIONS: (1) Irradiating fully synthesized NDDP is feasible for diagnostic use if a purification step is taken after the irradiation, and (2) radiation exposure is lessened by irradiating NDDP after synthesis rather than starting with high-specific-activity isotopes.


Subject(s)
Antineoplastic Agents/chemistry , Neutron Activation Analysis , Organoplatinum Compounds/chemistry , Drug Stability
10.
Invest New Drugs ; 18(3): 243-5, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10958592

ABSTRACT

Twenty-four patients with pleural mesothelioma received 50 mg/m2 of Doxil every four weeks. At follow-up, the disease had stabilized in 43% percent of patients and had progressed in 57%. No objective responses were observed. Estimated median survival of all patients was 37 weeks. Major toxicities were erythrodysesthesia of hands and feet and myelosuppression. No cardiac toxicity was observed. We concluded that Doxil at this dosage and schedule is inactive against pleural mesothelioma.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/therapeutic use , Mesothelioma/drug therapy , Pleural Neoplasms/drug therapy , Aged , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Humans , Infusions, Intravenous , Male , Middle Aged
11.
Am J Vet Res ; 61(7): 791-5, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10895902

ABSTRACT

OBJECTIVE: To determine clinical response and toxic effects of cis-bis-neodecanoato-trans-R,R-1,2-diaminocyclohexane platinum (II) (L-NDDP) administered i.v. at escalating doses to cats with oral squamous cell carcinoma (SCC). ANIMALS: 18 cats with oral SCC. PROCEDURE: Cats that failed to respond to conventional treatment or had nonresectable tumors were included. Data included a CBC, serum biochemical analyses, urinalysis, cytologic examination of a fine-needle aspirate of enlarged lymph nodes, and thoracic and oral radiographs for clinical staging. A starting dose (75 to 100 mg/m2 of L-NDDP) was administered i.v.. At 21-day intervals, subsequent doses increased by the rate of 5 or 10 mg/m2. Response was evaluated every 21 days by tumor measurement and thoracic radiography. Quality of life was assessed by owners, using a performance status questionnaire. RESULTS: On average, cats received 2 treatments. Toxicoses included an intermittent, acute anaphylactoid-parasympathomimetic reaction, lethargy or sedation (< or = 24 hours), inappetence or signs of depression (< or = 72 hours), mild to moderate increase in hepatic enzyme activity, and melena. Pulmonary, renal, or hematopoietic abnormalities were not evident. Performance status surveys indicated normal behavior and grooming or decreased activity and self-care (19/20 assessments), ate well with or without assistance (15/20), and did not lose weight (15/20). Median survival time was 59.8 days (mean, 54.1 days). CONCLUSIONS AND CLINICAL RELEVANCE: L-NDDP was ineffective for treatment of cats with oral SCC. None of the cats had a complete or partial remission. Acute toxicoses and poor therapeutic response limit therapeutic usefulness of L-NDDP in cats, unless dosage, frequency, and administration procedures can be improved.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/veterinary , Cat Diseases/drug therapy , Mouth Neoplasms/veterinary , Organoplatinum Compounds/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/drug therapy , Cat Diseases/diagnostic imaging , Cats , Female , Infusions, Intravenous/veterinary , Male , Mouth Neoplasms/diagnostic imaging , Mouth Neoplasms/drug therapy , Neoplasm Staging/veterinary , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Proportional Hazards Models , Prospective Studies , Radiography, Thoracic/veterinary , Surveys and Questionnaires
12.
Cancer Gene Ther ; 7(5): 683-96, 2000 May.
Article in English | MEDLINE | ID: mdl-10830716

ABSTRACT

Regional (intratracheal or aerosol) delivery of cationic liposome-DNA complexes for gene therapy of lung disease offers distinct advantages over systemic (intravenous) administration. However, optimal formulations for early lung cancer treatment have not been established. Therefore, we investigated >50 different liposome and micelle formulations for factors that may affect their transcription efficiency and tested the ideal formulations in an in vivo mouse model. Our data showed that cationic liposomes were generally more effective at transfecting genes than were micelles of the same lipid composition, thus suggesting a role for the bilayer structure in facilitating transfection. In addition, the transfection efficiency of liposome-delivered genes was highly dependent upon the lipid composition, lipid/DNA ratio, particle size of the liposome-DNA complex, and cell lines used. By optimizing these factors in vitro and in vivo, we developed a novel liposome formulation (DP3) suitable for intratracheal administration. Using G67 liposome as control, we found that DP3 was more effective than G67 in vitro and as effective as G67 at both preventing lung tumor growth and prolonging survival in our lung cancer mouse model. We observed a positive correlation between the in vitro p53 function and the in vivo antitumoral activities of liposome-p53 formulations, which had not been reported previously in studies of an intravenous liposome gene delivery system. This correlation may facilitate the development and optimization of a liposome-p53 formulation for aerosol use in lung cancer patients.


Subject(s)
Cations , Genes, p53/genetics , Genetic Therapy/methods , Liposomes/therapeutic use , Lung Neoplasms/therapy , Aerosols/therapeutic use , Animals , Apoptosis , DNA/pharmacology , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Micelles , Neoplasm Transplantation , Neoplasms, Experimental/therapy , Plasmids/genetics , Point Mutation , Time Factors , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
13.
J Microencapsul ; 17(3): 307-22, 2000.
Article in English | MEDLINE | ID: mdl-10819419

ABSTRACT

L-NDDP is a liposome-entrapped platinum compound currently in phase 2 clinical trials that has been shown to undergo intraliposomal activation. The degradation/activation kinetics of liposome entrapped cis-bis-neodecanoato-trans-R,R-1,2-diamminocyclohexane platinum (II) [L-NDDP] at different conditions of pH, and temperature is presented. Liposomes were reconstituted in a solution of 0.9% sodium chloride (NaCl) in water (pH 5) at room temperature (formulation conditions currently used in the ongoing clinical trials). In the temperature experiments, L-NDDP 0.9% sodium chloride liposomes were incubated in a water-bath at 40, 60, and 80 degrees C. In the pH experiments, these solutions were compared to water, phosphate with and without chloride ion present, phosphate buffer without chloride ion at pH 3.1, 5.0, and 7.4, and glycine buffer with and without chloride ion. In 0.9% sodium chloride at room temperature, the chemical degradation/activation of liposome-bound NDDP was biphasic, with most of the degradation (approximately 45% conversion) occurring during the first hour after formation of the liposome suspension. NDDP degradation was pH dependent: when using pH 3 phosphate buffer as a reconstituting solution, liposome-bound NDDP degraded rapidly, whereas in pH 7.4 phosphate buffer it was stable for > 72 h. NDDP degradation was also temperature-dependent, the 50% point decreasing from 12 h at 25 degrees C to 9.5 h at 40 degrees C, 3.8 h at 60 degrees C, and 0.3 h at 80 degrees C when using 0.9% NaCl in water as a reconstituting solution. Using glycine buffer solution with and without NaCl at room temperature, no NDDP degradation over a 72 h period was observed at 25 degrees C; however, at 40 degrees C, only 68% NDDP remained intact at 72 h. Atomic absorption spectrophotometry (AAS) analysis of the eluting fractions after injection of L-NDDP samples reconstituted in chloride-containing and non chloride-containing solutions clearly indicated that the formation of DACH-Pt-Cl2 was only observed when chloride-containing solutions were used and was first detected at 3 h when using 0.9% NaCl in water as a reconstituting solution. These results indicate that pH and temperature, and not the presence of chloride ion, are the main factors leading to the activation of NDDP. Since 45% of NDDP is already degraded at 1 h in the same conditions, it is concluded that (1) the first active intermediates of L-NDDP formed within the liposomes are the DACH-Pt chloro-aquo and diaquo intermediates, and (2) the in vivo, antitumour activity of L-NDDP is most likely mediated by direct intracellular delivery of the active species.


Subject(s)
Antineoplastic Agents/chemistry , Organoplatinum Compounds/chemistry , Antineoplastic Agents/administration & dosage , Buffers , Chlorides/chemistry , Drug Carriers , Drug Stability , Hydrogen-Ion Concentration , Kinetics , Liposomes , Organoplatinum Compounds/administration & dosage , Platinum/analysis , Powders , Spectrophotometry, Ultraviolet , Suspensions , Temperature
14.
Cancer Gene Ther ; 7(4): 530-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10811470

ABSTRACT

The immune responses of 10 patients with advanced non-small cell lung cancer receiving monthly intratumoral injections of a recombinant adenovirus containing human wild-type p53 (Ad-p53) to adenovirus and transgene antigens were studied. The predominate cellular and humoral immune responses as measured by lymphocyte proliferation and neutralizing antibody (Ab) formation were to adenovirus serotype 5 vector antigens, with increased responses in posttreatment samples. Consistent alterations in posttreatment cellular and humoral immune responses to p53 epitopes were not observed, and cytotoxic Abs to human lung cancer cells were not generated. Patients in this study had evidence of an antitumoral effect of this treatment with prolonged tumor stability or regression; however, neither Abs to p53 protein nor increased lymphocyte proliferative responses to wild-type or mutant p53 peptides have been consistently detected.


Subject(s)
Adenoviridae/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Genetic Therapy/methods , Lung Neoplasms/therapy , Tumor Suppressor Protein p53/immunology , Adenoviridae/genetics , Aged , Amino Acid Sequence , Antibody Formation , Carcinoma, Non-Small-Cell Lung/immunology , Cytotoxicity, Immunologic , Female , Gene Transfer Techniques , Genetic Vectors/immunology , Humans , Immunity, Cellular , Lung Neoplasms/immunology , Lymphocyte Activation , Male , Middle Aged , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/immunology , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
15.
Anticancer Res ; 20(2A): 693-702, 2000.
Article in English | MEDLINE | ID: mdl-10810342

ABSTRACT

The p53 gene plays an important role in the regulation of cell-cycle progression and apoptosis. Recent studies have implicated p53 in determining cell fate, and shown that p53 status is associated with cellular sensitivity to anticancer agents. However, the role of p53 in paclitaxel-induced cytotoxicity remains unclear. Here we show that the induction of exogenous wild-type (wt) p53 genes in p53-null human NSCLC H358 cells via transient gene transfection with cationic liposome-wt p53 complexes resulted in a typical senescence-like phenotype. In short, cell growth was reduced, homeostasis occurred, cell morphology became enlarged and flat, the cell cycle was arrested at G1 phase, cyclin B1 and cdc2 expression was down-regulated, and DNA synthesis was suppressed. The sensitivity of wt p53-transfected cells (H358/p53) to paclitaxel was approximately 3-fold lower than that of H358 cells. Paclitaxel treatment gradually and significantly blocked cell-cycle progression at G2/M phase and increased the accumulation of cyclin B1 and cdc2 in H358 cells. In contrast, the same treatment slightly arrested the cell cycle at G2/M phase and slightly elevated cyclin B1 expression in H358/p53 cells. The rate of uptake and efflux of paclitaxel was not significantly different between H358 and H358/p53 cells, indicating that the reduction in cellular sensitivity caused by p53 transfection was not due to alterasion in intracellular drug concentration. Together, our findings suggest that the induction of exogenous wt p53 gene expression in cells lacking p53 function can trigger the senescence program and that loss of sensitivity to paclitaxel by p53-transfected cells may be associated, at least in part, with the induction of a senescence-like phenotype.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cellular Senescence/genetics , Genes, p53 , Lung Neoplasms/pathology , Paclitaxel/toxicity , Tumor Suppressor Protein p53/metabolism , Biological Transport , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Survival/drug effects , Cell Survival/genetics , Humans , Kinetics , Lung Neoplasms/genetics , Paclitaxel/pharmacokinetics , Phenotype , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
16.
J Clin Oncol ; 18(3): 609-22, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10653876

ABSTRACT

PURPOSE: To determine the safety and tolerability of adenovirus-mediated p53 (Adp53) gene transfer in sequence with cisplatin when given by intratumor injection in patients with non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: Patients with advanced NSCLC and abnormal p53 function were enrolled onto cohorts receiving escalating dose levels of Adp53 (1 x 10(6) to 1 x 10(11) plaque-forming units [PFU]). Patients were administered intravenous cisplatin 80 mg/m(2) on day 1 and study vector on day 4 for a total of up to six courses (28 days per course). Apoptosis was determined by the terminal deoxynucleotidyl- transferase-dUTP nick-end labeling assay. Evidence of vector-specific sequences were determined using reverse-transcriptase polymerase chain reaction. Vector dissemination and biodistribution was monitored using a series of assays (cytopathic effects assay, Ad5 hexon enzyme-linked immunosorbent assay, vector-specific polymerase chain reaction assay, and antibody response assay). RESULTS: Twenty-four patients (median age, 64 years) received a total of 83 intratumor injections with Adp53. The maximum dose administered was 1 x 10(11) PFU per dose. Transient fever related to Adp53 injection developed in eight of 24 patients. Seventeen patients achieved a best clinical response of stable disease, two patients achieved a partial response, four patients had progressive disease, and one patient was not assessable. A mean apoptotic index between baseline and follow-up measurements increased from 0.010 to 0.044 (P =.011). Intratumor transgene mRNA was identified in 43% of assessable patients. CONCLUSION: Intratumoral injection with Adp53 in combination with cisplatin is well tolerated, and there is evidence of clinical activity.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/therapy , Cisplatin/therapeutic use , Gene Transfer Techniques , Genes, p53 , Lung Neoplasms/drug therapy , Lung Neoplasms/therapy , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Adult , Aged , Antibodies, Viral/biosynthesis , Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/adverse effects , Combined Modality Therapy , DNA Mutational Analysis , DNA, Neoplasm/genetics , Female , Gene Transfer Techniques/adverse effects , Genetic Vectors/genetics , Humans , In Situ Nick-End Labeling , Injections, Intralesional , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Organ Specificity/genetics , Staining and Labeling
17.
Clin Cancer Res ; 6(1): 193-202, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10656450

ABSTRACT

Cell cycle regulators and signal transduction pathways can influence apoptotic sensitivity of tumor cells, and we previously described an association between EGFr overexpression, reduced DNA repair activity, and increased apoptotic sensitivity of ME-180 cervical carcinoma cells toward cis-diammedichloroplatinum (cDDP; K. Nishikawa, et al., Cancer Res., 52: 4758-4765, 1992). In the present study, the characteristics of ME-180 cells selected for high or low apoptotic sensitivity to cDDP (or camptothecin) were examined and compared to determine whether signal transduction components and cell cycle regulation were distinct in these isogenic drug response variant populations. As ME-180 cells progressed from high to low cDDP sensitivity [IC50 approximately 80 ng/ml in cDDP sensitive (PT-S) to approximately 2000 ng/ml in cDDP-resistant (Pt-R) cells], there was a significant decrease in EGFr expression that paralleled the relative reduction in cDDP apoptotic responsiveness (approximately 30-fold). cDDP-resistant cells had the slowest rate of growth and more effectively reduced DNA adduct levels following cDDP exposure than parental cells. Cellular levels of the cell cycle inhibitor p21WAF1 inversely correlated with cDDP responsiveness with high levels of p21WAF1 expressed in drug-resistant Pt-R cells in the absence of elevated p53. cDDP stimulated a 2-fold increase in p53 levels in both drug-sensitive and drug-resistant cells but caused a delayed reduction in p21WAF1 levels, suggesting p53-independent regulation of p21WAF1 in ME-180 cells. Activation of EGFr in Pt-R cells stimulated cell cycle progression (2-fold), reduced p21WAF1 levels (>2-fold), and increased sensitivity to cDDP (3-fold), suggesting that receptor signaling enhanced the efficacy of cDDP to induce cell death by relieving cell cycle restriction. These results demonstrate that the transition of ME-180 cells from a drug-sensitive to drug-resistant phenotype correlates with reciprocal changes in EGFr and p21WAF1 expression and provides additional evidence that the pathways controlled by these proteins may contribute to some forms of drug resistance.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Cisplatin/toxicity , Cyclins/physiology , ErbB Receptors/physiology , Cell Division/drug effects , Cisplatin/pharmacokinetics , Cyclin-Dependent Kinase Inhibitor p21 , DNA Adducts , Drug Resistance, Neoplasm , Enzyme Inhibitors/metabolism , Epidermal Growth Factor/pharmacology , Female , Humans , Tumor Cells, Cultured , Uterine Cervical Neoplasms
18.
Clin Cancer Res ; 6(12): 4932-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11156254

ABSTRACT

The lack of tumor models that can reliably predict for response to anticancer agents remains a major deficiency in the field of experimental cancer therapy. Although heterotransplants of certain human solid tumors can be successfully grown in nude mice, they have never been appropriately explored for prediction of in vivo chemosensitivity to anticancer agents. We determined the tumor response rate and studied the influence of several biological and molecular tumor parameters on the in vivo sensitivity to paclitaxel in a series of heterotransplanted human non-small cell lung cancer (NSCLC) tumors. One hundred consecutive resected NSCLC tumors were heterotransplanted s.c. in nude mice. The in vivo sensitivity to i.v. paclitaxel (60 mg/kg every 3 weeks) was studied in 34 successfully grown heterotransplants. Treatment started when the tumors reached a size of 5 mm in diameter, and strict standard clinical criteria (>50% shrinkage in tumor weight or cross-sectional surface) were used to define tumor response. Baseline multidrug resistance protein (MRP), Her-2/neu, and epidermal growth factor receptor (EGFR) expression, and pre- and posttherapy bax and bcl-2 expression were determined by Western blot analysis. p53 status was determined by sequencing. The overall take rate was 46% (95% confidence interval, 36-56%) and was significantly higher (P < 0.05) for squamous carcinoma tumors (75%) than for adenocarcinoma tumors (30%) and bronchoalveolar tumors (23%). The heterotransplants were morphologically very similar to the original tumors. The response rate to paclitaxel was 21% (95% confidence interval, 9-38%). Baseline tumor parameters associated with response were no Her-2/neu expression (none of the responding tumors expressed Her-2/neu versus 48% of the nonresponding tumors, P = 0.05) and baseline bcl-2 expression (all responding tumors expressed bcl-2 versus only 43% of the nonresponding tumors, P = 0.02). There was a trend toward a higher response rate in bax-positive tumors, and MRP- and EGFR-negative tumors, but it was not statistically significant. The response was independent of baseline p53 status and baseline mitotic index. Responding tumors had a higher bax/bcl-2 ratio 24 h after therapy, but the difference was only marginally significant (2.8 for responding tumors versus 1.1 for nonresponding tumors, P = 0.07). The extent of mitotic arrest at 24 h after therapy was not associated with response. Human NSCLC heterotransplants are morphologically identical to the original tumors and have a response rate to paclitaxel that is equivalent to that reported in Phase II studies in patients with advanced NSCLC treated with single-agent paclitaxel. NSCLC heterotransplants deserve to be explored to evaluate new agents for lung cancer and to predict clinical response on an individual basis in selected groups of patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , ATP-Binding Cassette Transporters/biosynthesis , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Blotting, Western , Bronchial Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , ErbB Receptors/biosynthesis , Genes, p53/genetics , Humans , Mice , Mice, Nude , Mitosis/drug effects , Multidrug Resistance-Associated Proteins , Mutation , Neoplasm Transplantation , Polymerase Chain Reaction , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptor, ErbB-2/biosynthesis , Time Factors , Tumor Cells, Cultured , bcl-2-Associated X Protein
19.
J Clin Oncol ; 17(8): 2309-15, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10561292

ABSTRACT

PURPOSE: The combination of cisplatin, etoposide, and paclitaxel was studied in patients with extensive small-cell lung cancer in a phase I component followed by a phase II trial to determine the maximum-tolerated dose (MTD), characterize toxicity, and estimate response and median survival rates. PATIENTS AND METHODS: Forty-one patients were treated between October 1993 and April 1997. Doses for the initial cohort were cisplatin 75 mg/m(2) on day 1, etoposide 80 mg/m(2)/d on days 1 to 3, and paclitaxel 130 mg/m(2) on day 1 over 3 hours. Cycles were repeated every 3 weeks for up to six cycles. The MTD was reached in the first six patients. In these six patients and in the next 35 patients, who were entered onto the phase II trial, response and survival were estimated. RESULTS: At the initial dose level, one of six patients developed febrile neutropenia, and five of six achieved targeted neutropenia (nadir absolute granulocyte count, 100 to 1,000/microL) without any other dose-limiting toxicity, defining this level as the MTD. Grade 4 neutropenia was observed in 88 (47%) of 188 total courses administered at or less than the MTD. Neutropenia was associated with fever in only 17 (9%) of 188 courses, but two patients experienced neutropenic sepsis that was fatal. Nonhematologic toxicity greater than grade 2 was observed in 10 (5%) of 188 total courses, with fatigue, peripheral neuropathy, and nausea/vomiting most common. The overall objective response rate was 90% of 38 assessable patients: six complete responses (16%) and 28 partial responses(74%). Median progression-free and overall survival durations were 31 and 47 weeks, respectively. CONCLUSION: The combination of cisplatin, etoposide, and paclitaxel produced response and survival rates similar to those of other combinations and was well tolerated.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Small Cell/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Carcinoma, Small Cell/mortality , Carcinoma, Small Cell/pathology , Cisplatin/administration & dosage , Disease-Free Survival , Drug Administration Schedule , Etoposide/administration & dosage , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Neutropenia/chemically induced , Paclitaxel/administration & dosage
20.
J Natl Cancer Inst ; 91(9): 763-71, 1999 May 05.
Article in English | MEDLINE | ID: mdl-10328106

ABSTRACT

BACKGROUND: Preclinical studies in animal models have demonstrated tumor regression following intratumoral administration of an adenovirus vector containing wild-type p53 complementary DNA (Ad-p53). Therefore, in a phase I clinical trial, we administered Ad-p53 to 28 patients with non-small-cell lung cancer (NSCLC) whose cancers had progressed on conventional treatments. METHODS: Patients received up to six, monthly intratumoral injections of Ad-p53 by use of computed tomography-guided percutaneous fine-needle injection (23 patients) or bronchoscopy (five patients). The doses ranged from 10(6) plaque-forming units (PFU) to 10(11) PFU. RESULTS: Polymerase chain reaction (PCR) analysis showed the presence of adenovirus vector DNA in 18 (86%) of 21 patients with evaluable posttreatment biopsy specimens; vector-specific p53 messenger RNA was detected by means of reverse transcription-PCR analysis in 12 (46%) of 26 patients. Apoptosis (programmed cell death) was demonstrated by increased terminal deoxynucleotide transferase-mediated biotin uridine triphosphate nick-end labeling (TUNEL) staining in posttreatment biopsy specimens from 11 patients. Vector-related toxicity was minimal (National Cancer Institute's Common Toxicity Criteria: grade 3 = one patient; grade 4 = no patients) in 84 courses of treatment, despite repeated injections (up to six) in 23 patients. Therapeutic activity in 25 evaluable patients included partial responses in two patients (8%) and disease stabilization (range, 2-14 months) in 16 patients (64%); the remaining seven patients (28%) exhibited disease progression. CONCLUSIONS: Repeated intratumoral injections of Ad-p53 appear to be well tolerated, result in transgene expression of wild-type p53, and seem to mediate antitumor activity in a subset of patients with advanced NSCLC.


Subject(s)
Adenoviridae , Carcinoma, Non-Small-Cell Lung/therapy , Gene Transfer Techniques , Genes, p53 , Genetic Therapy/methods , Lung Neoplasms/therapy , Adenoviridae/genetics , Adult , Aged , Bronchoscopy , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , DNA, Viral/isolation & purification , Disease Progression , Female , Genes, p53/genetics , Genetic Vectors/adverse effects , Humans , In Situ Nick-End Labeling , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Patient Selection , Survival Analysis , Tomography, X-Ray Computed , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...