Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Cancer Chemother Pharmacol ; 72(5): 1073-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24048674

ABSTRACT

PURPOSE: The potential synergy of modulating platinum-induced DNA damage by combining the proteasome inhibitor bortezomib with oxaliplatin was studied in patients with solid tumors, with special attention to avoidance of cumulative neurotoxicity (NT). PATIENTS AND METHODS: In a 3 + 3 dose escalation design, patients received bortezomib at 1.0-1.5 mg/m² on days 1 and 4 and oxaliplatin at 60-85 mg/m² on day 1 of a 14-day cycle. NT assessments were performed at the start of every two cycles. Oxaliplatin pharmacokinetics (PK) were determined pre- and post-bortezomib. RESULTS: Thirty patients were enrolled with 25 (11 men, 14 women) fully evaluable for NT assessments at cycle 2. The median age was 56 years (range 35-74 years); median number of cycles received 2 (range 1-10). At dose levels 2-5 (B 1.3 mg/m²), patients manifested NT grades 3 and 4 at a median 3.4 cycles (range 2-9 cycles): 3 had ataxia (one also with sensory neuropathy or neurogenic hypotension, respectively) and 3 had just sensory neuropathy. A 6th dose-level reducing bortezomib to 1.0 mg/m² with oxaliplatin 85 mg/m²) was explored and no NT or dose limiting toxicities were noted among 7 evaluable patients (5 receiving two or more cycles). Four patients experienced a partial response--one with platinum-resistant ovarian cancer, another with gastroesophageal cancer, another with ampulla of Vater carcinoma, and a patient with cholangiocarcinoma. PK studies at dose levels 1 and 2 showed greater mean ultrafiltrable platinum when oxaliplatin was dosed after bortezomib. CONCLUSIONS: Bortezomib 1.0 mg/m² × 2 every 14 days combines safely with oxaliplatin. At higher doses, cumulative NT (i.e., cerebellar signs and sensory neuropathy) occurs at an accelerated pace perhaps from a PK interaction.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/therapeutic use , Neoplasms/drug therapy , Neurotoxicity Syndromes/prevention & control , Organoplatinum Compounds/therapeutic use , Proteasome Inhibitors/therapeutic use , Pyrazines/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Bortezomib , Cohort Studies , Dose-Response Relationship, Drug , Drug Monitoring , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Humans , Male , Middle Aged , Neoplasms/blood , Neoplasms/metabolism , Neoplasms/pathology , Neurotoxicity Syndromes/physiopathology , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/pharmacokinetics , Ovarian Neoplasms/blood , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Oxaliplatin , Proteasome Inhibitors/administration & dosage , Proteasome Inhibitors/adverse effects , Pyrazines/administration & dosage , Pyrazines/adverse effects , Severity of Illness Index , Tumor Burden/drug effects
2.
Oncogene ; 27(22): 3201-10, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18193092

ABSTRACT

While epidermal growth factor receptor (EGFR) dysregulation is known to play a critical role in prostate carcinogenesis, there has been no direct evidence indicating EGFR mutations induce tumorigenesis in prostate cancer. We previously identified four novel EGFR somatic mutations in the EGFR tyrosine kinase domain of prostate cancer patients: G735S, G796S, E804G and R841K. In this study, we investigated the oncogenic potential of these somatic mutations by establishing stable clonal NIH3T3 cells expressing these four mutations and WT EGFR to determine their ability to increase cell proliferation and invasion. In the absence of the EGF ligand, cell proliferation was readily increased in G735S, G796S and E804G mutants compared to WT EGFR. The addition of EGF ligand greatly increased cell growth and transforming ability of these same EGFR mutants. Matrigel invasion assays showed enhanced invasion with G735S, G796S and E804G mutants. Western blot analysis showed that these EGFR mutations enhanced cell growth and invasion via constitutive and hyperactive tyrosine phosphorylation and led to the activation of mitogen-activated protein kinase (MAPK), signal transducer and activator of transcription 3 (STAT3) and Akt pathways. Our findings demonstrate the oncogenic activation of three novel EGFR somatic missense mutations in prostate cancer. Molecules that regulate the mechanisms of their oncogenic activation represent novel targets for limiting tumor cell progression, and further elucidation of these mutations will have utility in prostate cancer treatment.


Subject(s)
ErbB Receptors/genetics , ErbB Receptors/physiology , Mutation, Missense , Prostatic Neoplasms/genetics , Animals , Base Sequence , Cell Movement/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cloning, Molecular , DNA Mutational Analysis , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mutagenesis, Site-Directed , NIH 3T3 Cells , Prostatic Neoplasms/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Structure, Tertiary/genetics , Signal Transduction/genetics , Tumor Stem Cell Assay
3.
Gynecol Oncol ; 103(3): 783-92, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17070570

ABSTRACT

Ovarian cancer is the leading cause of gynecologic cancer deaths in the U.S. The concept of intraperitoneal drug delivery for therapy of intraperitoneal cancers, such as ovarian cancer, arose in the 1960s. The field of intraperitoneal cisplatin therapy for ovarian cancer was initiated in the late 1970s and early 1980s. The markedly improved survival data resulting from a phase III trial of intraperitoneal cisplatin for ovarian cancer in early 2006 led to an NCI Clinical Announcement and a Gynecologic Oncology Group-sponsored workshop on intraperitoneal therapy in January, 2006, in San Diego, California. The proceedings of this workshop summarize both research trial results and practical implementation issues associated with intraperitoneal therapy discussed at this workshop.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/administration & dosage , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , Congresses as Topic , Disease-Free Survival , Female , Humans , Injections, Intraperitoneal , Ovarian Neoplasms/mortality , Ovarian Neoplasms/nursing , Randomized Controlled Trials as Topic , Survival Analysis , United States
4.
J Clin Oncol ; 23(25): 6107-16, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16135477

ABSTRACT

PURPOSE: We performed a phase I study of a day (D) 1 and D4 bortezomib administration once every 2 weeks to determine the recommended phase II dose and toxicity profile, and the extent of 20S proteasome inhibition obtained. PATIENTS AND METHODS: Patients with solid tumors or lymphomas were treated with bortezomib at 0.25 to 1.9 mg/m2 on D1 and D4, every 2 weeks. 20S proteasome levels in blood were assayed at baseline and at 1, 4, and 24 hours postdose in cycle 1. RESULTS: On this D1 and D4 every 2 weeks' schedule, dose-limiting toxicity (DLT) was evident at the 1.75 and 1.9 mg/m2 dose levels, most commonly in patients receiving individual total doses > or = 3.0 mg. The main DLT was peripheral neuropathy evident at the higher doses and in patients previously exposed to neurotoxic agents. Other DLTs included diarrhea and fatigue; grade 3 thrombocytopenia was also noted. Reversible inhibition of 20S proteasome activity was dose dependent and best fit a total dose (mg) per fraction rather than mg/m2; 70% of baseline activity was inhibited by a dose of 3.0 to 3.5 mg given on D1 and on D4 every other week. Antitumor effects short of confirmed partial responses were observed in patients with melanoma, non-small-cell lung cancer, and renal cell carcinoma. CONCLUSION: Bortezomib (PS-341) is a novel antineoplastic agent that is well tolerated at doses not exceeding 3.0 mg (equivalent to 1.75 mg/m2), repeated on D1 and D4 every other week. This dose correlates with 70% inhibition of 20S proteasome activity. DLTs include neuropathy, fatigue, and diarrhea.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Boronic Acids/pharmacology , Boronic Acids/pharmacokinetics , Pyrazines/pharmacology , Pyrazines/pharmacokinetics , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Boronic Acids/administration & dosage , Boronic Acids/therapeutic use , Bortezomib , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Lymphoma/drug therapy , Male , Middle Aged , Neoplasms/drug therapy , Peripheral Nervous System/drug effects , Peripheral Nervous System/pathology , Proteasome Endopeptidase Complex/blood , Proteasome Inhibitors , Pyrazines/administration & dosage , Pyrazines/therapeutic use , Treatment Outcome
5.
Ann Oncol ; 14(9): 1430-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12954584

ABSTRACT

BACKGROUND: Pegylated liposomal doxorubicin (Doxil) has been reported to cause immediate hypersensitivity reactions (HSRs) that cannot be explained as IgE-mediated (type I) allergy. Previous in vitro and animal studies indicated that activation of the complement (C) system might play a causal role in the process, a proposal that has not been tested in humans to date. PATIENTS AND METHODS: Patients with solid tumors (n = 29) treated for the first time with Doxil were evaluated for HSRs and concurrent C activation. HSRs were classified from mild to severe, while C activation was estimated by serial measurement of plasma C terminal complex (SC5b-9) levels. Increases in SC5b-9 were compared in patients with or without reactions, and were correlated with Doxil dose rate. RESULTS: Moderate to severe HSRs occurred in 45% of patients. Plasma SC5b-9 at 10 min after infusion was significantly elevated in 92% of reactor patients versus 56% in the non-reactor group, and the rise was greater in reactors than in non-reactors. We found significant association between C activation and HSRs, both showing direct correlation with the initial Doxil dose rate. CONCLUSIONS: C activation may play a key role in HSRs to Doxil. However, low-level C activation does not necessarily entail clinical symptoms, highlighting the probable involvement of further, as yet unidentified, amplification factors.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Complement Activation/drug effects , Doxorubicin/adverse effects , Drug Hypersensitivity/immunology , Hypersensitivity, Immediate/immunology , Adult , Aged , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasms/drug therapy
6.
Melanoma Res ; 12(1): 83-90, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11828262

ABSTRACT

We evaluated the effects of vitamin E (dl-alpha-tocopherol) on mutagen sensitivity levels in a randomized placebo-controlled pilot trial. In brief, a dietary supplement of 1000 mg/day vitamin E or a placebo was randomly administered for 3 months to melanoma outpatients clinically free of the disease. Plasma vitamin E and mutagen sensitivity levels were measured at baseline and at the end of the trial after 3 months. At baseline, we found no significant differences in plasma vitamin E and mutagen sensitivity levels between the two groups. We also measured dietary intake at baseline and found dietary vitamin E to be a poor predictor of plasma levels of vitamin E. After 3 months of supplementation, we found that plasma levels of alpha-tocopherol increased significantly (P = 0.0005) in the vitamin E compared to the placebo group. We also found a non-significant, but consistent decrease in plasma gamma-tocopherol concentrations in the vitamin E supplemented compared to the placebo group. We did not find any significant difference between the vitamin E and placebo groups in mutagen sensitivity levels either at baseline or after 3 months of supplementation. We conclude that short term vitamin E supplementation, although it causes increased blood levels of alpha-tocopherol, does not provide protection against bleomycin-induced chromosome damage.


Subject(s)
Melanoma/metabolism , Mutagens , alpha-Tocopherol/therapeutic use , Adult , Aged , Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Cells, Cultured , Chromosomes/drug effects , Dietary Supplements , Female , Humans , Lymphocytes/metabolism , Male , Middle Aged , Pilot Projects , Placebos , Vitamin E/blood , gamma-Tocopherol/blood
7.
J Liposome Res ; 12(1-2): 165-72, 2002.
Article in English | MEDLINE | ID: mdl-12604051

ABSTRACT

Pegylated liposomal doxorubicin (Doxil) and 99mTc-HYNIC PEG liposomes (HPL) were reported earlier to cause hypersensitivity reactions (HSRs) in a substantial percentage of patients treated i.v. with these formulations. Here we report that (1) Doxil, HPL, pegylated phosphatidylethanolamine (PEG-PE)-containing empty liposomes matched with Doxil and HPL in size and lipid composition, and phosphatidylglycerol (PG)-containing negatively charged vesicles were potent C activators in human serum in vitro, whereas small neutral liposomes caused no C activation. (2) Doxil and other size-matched PEG-PE and/or PG-containing liposomes also caused massive cardiopulmonary distress with anaphylactoid shock in pigs via C activation, whereas equivalent neutral liposomes caused no hemodynamic changes. (3) A clinical study showed more frequent and greater C activation in patients displaying HSR than in non-reactive patients. These data suggest that liposome-induced HSRs in susceptible individuals may be due to C activation, which, in turn, is due to the presence of negatively charged PEG-PE in these vesicles.


Subject(s)
Complement Activation , Liposomes/metabolism , Polyethylene Glycols/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Humans , Lipid Metabolism , Neoplasms/metabolism , Polyethylene Glycols/pharmacology , Swine , Technetium/pharmacology , Time Factors
8.
Anal Biochem ; 291(2): 279-89, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11401302

ABSTRACT

Dietary and pharmacologic isothiocyanates (ITCs) may play a role in reducing the risk of certain cancers. The quantification of ITCs in humans is important both for epidemiological and pharmacokinetic studies. We describe a modification of an HPLC-based assay of urinary ITCs for use with human plasma. The assay utilizes the cyclocondensation reaction of 1,2-benzenedithiol with ITCs present in human plasma, followed by a two-step hexane extraction and analysis by HPLC using UV detection at 365 nm. The method shows linearity and reproducibility with human plasma over a range of 49-3003 nM phenethyl isothiocyanate (PEITC) (r(2) = 0.996 +/- 0.003). A similar degree of linearity was seen with two other biologically occurring conjugates of PEITC: PEITC--N-acetylcysteine (PEITC--NAC) and PEITC--glutathione (PEITC--GSH). The recovery of PEITC assessed on multiple days was 96.6 +/- 1.5% and was 100% for PEITC--GSH and PEITC--NAC. The reproducibility of the assay on multiday samplings showed a mean %CV of 6.5 +/- 0.3% for PEITC, 6.4 +/- 4.3 for PEITC--NAC and 12.3 +/- 3.9 for PEITC--GSH. In clinical studies, mean plasma ITC level of 413 +/- 193 nM PEITC equivalents was determined for a non-dietary-controlled group of 23 subjects. Multiday analysis data from pharmacokinetic plasma sets of 3 subjects taking a single dose of PEITC at 40 mg showed a good CV (range: 16-21%). The applicability of the methodology to pharmacokinetic studies of PEITC in humans is demonstrated.


Subject(s)
Isothiocyanates/blood , Chromatography, High Pressure Liquid , Clinical Trials, Phase I as Topic , Diet , Humans , Isothiocyanates/chemistry , Isothiocyanates/pharmacokinetics , Regression Analysis , Reproducibility of Results , Sensitivity and Specificity
9.
Cancer Res ; 61(7): 2996-3001, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306478

ABSTRACT

Clinical radioimmunotherapy (RIT) of solid tumors holds great promise, but as yet has been unable to deliver tumoricidal radiation doses without unacceptable toxicity. Our experimental approach aims to potentiate the therapeutic action of radioimmunoconjugates at the tumor site and thus improve the efficacy of RIT by combination with other treatment modalities. The topoisomerase I inhibitors are a unique class of chemotherapeutic agents that interfere with DNA breakage-reunion by inhibiting the action of topoisomerase I. Preclinical studies suggest that prolonged infusion of topoisomerase I inhibitors enhances cell toxicity due to ionizing radiation. We evaluated the efficacy of combined treatment with continuous administration of topotecan and 90Y-MX-DPTA BrE3 monoclonal antibody (which recognizes an epitope of breast epithelial mucin expressed in most breast cancers) on human mammary carcinoma xenografts in nude mice. Topotecan or 90Y-BrE3 treatment alone delayed overall tumor growth rate transiently but did not affect survival. The combination of RIT with topotecan substantially reduced growth of relatively large established tumors and caused complete tumor regressions and prolonged tumor-free survival in a substantial proportion of treated animals. In vitro studies demonstrated an increase in apoptotic rate and a decrease in cell proliferation of tumor cell lines treated with this combination. We combined the radiosensitization property of topotecan and the specificity of systemic RIT to establish a novel therapy for solid tumors in an experimental tumor xenograft model.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Immunotoxins/therapeutic use , Radioimmunotherapy , Topotecan/pharmacology , Yttrium Radioisotopes/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Bone Marrow/drug effects , Bone Marrow/radiation effects , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Division/radiation effects , Combined Modality Therapy , Drug Synergism , Enzyme Inhibitors/pharmacology , Female , Humans , Immunoglobulin G/immunology , Mice , Mice, Nude , Pentetic Acid/analogs & derivatives , Radiation Tolerance/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/administration & dosage
10.
Antimicrob Agents Chemother ; 45(2): 517-24, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158749

ABSTRACT

Hypericin is a natural derivative of the common St. Johns wort plant, Hypericum perforatum. It has in vitro activity against several viruses, including bovine diarrhea virus, a pestivirus with structural similarities to hepatitis C virus (HCV). We conducted a phase I dose escalation study to determine the safety and antiviral activity of hypericin in patients with chronic HCV infection. The first 12 patients received an 8-week course of 0.05 mg of hypericin per kg of body weight orally once a day; 7 patients received an 8-week course of 0.10 mg/kg orally once a day. At the end of the 8-week period of treatment, no subject had a change of plasma HCV RNA level of more than 1.0 log(10). Five of 12 subjects receiving the 0.05-mg/kg/day dosing schedule and 6 of 7 subjects receiving the 0.10-mg/kg/day dosing schedule developed phototoxic reactions. No other serious adverse events associated with hypericin use occurred. The pharmacokinetic data revealed a long elimination half-life (mean values of 36.1 and 33.8 h, respectively, for the doses of 0.05 and 0.1 mg/kg) and mean area under the curve determinations of 1.5 and 3.1 microg/ml x hr, respectively. In sum, hypericin given orally in doses of 0.05 and 0.10 mg/kg/d caused considerable phototoxicity and had no detectable anti-HCV activity in patients with chronic HCV infection.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Perylene/analogs & derivatives , Perylene/therapeutic use , Adolescent , Adult , Anthracenes , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Aspartate Aminotransferases/blood , Female , Follow-Up Studies , Half-Life , Hepacivirus/chemistry , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/virology , Humans , Male , Middle Aged , Perylene/adverse effects , Perylene/pharmacokinetics , Photosensitivity Disorders/chemically induced , RNA, Viral/analysis
11.
Photochem Photobiol ; 71(2): 188-95, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10687393

ABSTRACT

Photodynamically induced virus inactivation appears promising in preventing transmission of enveloped virus infections in transfusible blood products. The potential for utilizing hypericin as a photosensitizer to inactivate key enveloped viruses in packed red cell concentrates (PRC) was evaluated. In addition to inactivating effectively > or = 10(6) TCID50 of human immunodeficiency virus (HIV), inactivation of bovine viral diarrhea virus (BVDV) in PRC was used as a model for hepatitis C virus to overcome the deficiency in reliable experimental systems for hepatitis C virus (HCV) inactivation. BVDV was two orders of magnitude more sensitive to inactivation by hypericin than HIV. As part of the virucidal efficacy analyses, the effects of photosensitization on hemopoietic cell lines carrying quiescent integrated HIV provirus were studied as models for evaluating virus inactivation in latently infected cells. Phorbol ester-induced virus production by these cells was effectively prevented by photosensitization with hypericin. A refinement of the illumination conditions, incorporating a monochromatic sodium light source with an emission spectrum coinciding with the absorption peak of hypericin, was highly virucidal, however, caused unacceptable levels of hemolysis. Red blood cells could be protected from phototoxic cellular damage by complexing hypericin with human serum albumin (albumin-hypericin), but the decrease in hemolysis was at the expense of virucidal efficacy. Thus, excitation of hypericin with a fluorescent source appears to be useful potentially for virus inactivation in PRC.


Subject(s)
Antiviral Agents/pharmacology , Diarrhea Viruses, Bovine Viral/drug effects , Erythrocytes/virology , HIV-1/drug effects , Perylene/analogs & derivatives , Animals , Anthracenes , Anti-HIV Agents/pharmacology , Cattle , Cell-Free System , Diarrhea Viruses, Bovine Viral/radiation effects , HIV-1/radiation effects , Humans , Light , Perylene/pharmacology , Radiation-Sensitizing Agents/pharmacology
12.
Ann N Y Acad Sci ; 922: 178-87, 2000.
Article in English | MEDLINE | ID: mdl-11193893

ABSTRACT

The i.p. administration of topoisomerase I (Topo I) inhibitors has a pharmacologic advantage over intravenous application, including preservation of the biologically active lactone form. In our ongoing study, patients have received 9-amino-20(S)-camptothecin (9-AC) i.p. on days 1, 3, 5, 8, 10, and 12, repeated every 4 weeks. The daily dose has been escalated to level IV of 1.5 mg/m2 (9.0 mg/m2 per course), median of 3 cycles, range 1-4, with a reversible Grade 3 neutropenia in one patient. Responses included one CR (resolution of a pleural effusion), two patients without progressive disease (PD), two not evaluable, and two patients too early for evaluation. The area under the curve (AUC)i.p./AUCpl ratio (pharmacologic advantage) ranged from 7.6 to 16.5 on average, and, using nonlinear modeling, the pharmacologic decay data were fit to one- or two-compartmental models. Overall, a 9-AC i.p. application is well tolerated and anticipated to be an active regimen against i.p. malignancies, particularly those known to be sensitive to systemic Topo-I inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/administration & dosage , Enzyme Inhibitors/administration & dosage , Topoisomerase I Inhibitors , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Camptothecin/adverse effects , Camptothecin/pharmacokinetics , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Infusions, Parenteral , Neoplasms/drug therapy , Neoplasms/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism
13.
Nutr Cancer ; 38(2): 168-78, 2000.
Article in English | MEDLINE | ID: mdl-11525594

ABSTRACT

The cancer-chemopreventive effects of broccoli may be attributed, in part, to isothiocyanates (ITCs), hydrolysis products of glucosinolates. Glucosinolates are hydrolyzed to their respective ITCs by the enzyme myrosinase, which is inactivated by heat. In this study, the metabolic fate of glucosinolates after ingestion of steamed and fresh broccoli was compared in 12 male subjects in a crossover design. During each 48-hour baseline period, no foods containing glucosinolates or ITCs were allowed. The subjects then consumed 200 g of fresh or steamed broccoli; all other dietary sources of ITCs were excluded. Blood and urine samples were collected during the 24-hour period after broccoli consumption. Total ITC equivalents in broccoli and total ITC equivalents in plasma and urine were assayed by high-performance liquid chromatography as the cyclocondensation product of 1,2-benzenedithiol. The content of ITCs in fresh and steamed broccoli after myrosinase treatment was found to be virtually identical (1.1 vs. 1.0 micromol/g wet wt). The average 24-hour urinary excretion of ITC equivalents amounted to 32.3 +/- 12.7% and 10.2 +/- 5.9% of the amounts ingested for fresh and steamed broccoli, respectively. Approximately 40% of total ITC equivalents in urine, 25.8 +/- 13.9 and 6.9 +/- 2.5 micromol for fresh and steamed broccoli, respectively, occurred as the N-acetyl-L-cysteine conjugate of sulforaphane (SFN-NAC). Total ITC metabolites in plasma peaked between 0 and 8 hours, whereas urinary excretion of total ITC equivalents and SFN-NAC occurred primarily between 2 and 12 hours. Results of this study indicate that the bioavailability of ITCs from fresh broccoli is approximately three times greater than that from cooked broccoli, in which myrosinase is inactivated. Considering the cancer-chemopreventive potential of ITCs, cooking broccoli may markedly reduce its beneficial effects on health.


Subject(s)
Anticarcinogenic Agents/pharmacokinetics , Brassica/chemistry , Cooking , Glucose/analogs & derivatives , Glucosinolates/pharmacokinetics , Glycoside Hydrolases/metabolism , Thiocyanates/pharmacokinetics , Adult , Anticarcinogenic Agents/analysis , Biological Availability , Brassica/enzymology , Chromatography, High Pressure Liquid , Cross-Over Studies , Glucose/analysis , Glucosinolates/analysis , Humans , Imidoesters/analysis , Intestinal Absorption , Isothiocyanates , Male , Middle Aged , Oximes , Sulfoxides , Thiocyanates/analysis
14.
J Clin Oncol ; 17(8): 2553-61, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10561322

ABSTRACT

PURPOSE: Twenty-one-day topotecan infusion was administered as second-line therapy in patients with previously treated ovarian cancer (based on our prior favorable phase I experience) to determine its activity, time to progression, and pharmacodynamics. PATIENTS AND METHODS: Ovarian cancer patients with measurable lesions and one prior platinum-containing regimen were eligible. Topotecan 0.4 mg/m(2)/d 21-day continuous ambulatory intravenous infusion, with appropriate dose modifications for toxicity, was administered every 28 days. Weekly blood levels of topotecan and topoisomerase-1 (topo-1) levels in peripheral-blood mononuclear cells (PBMCs) were determined for pharmacodynamic correlation. RESULTS: Twenty-four patients were entered onto the study (six cisplatin-refractory, five relapsing within < 6 months and 13 relapsing > 6 months after platinum-based therapy). A total of 128 cycles of topotecan (median, four cycles per patient; range, one to 12 cycles) were administered. The major toxicity was neutropenia (29% grade 3 in all cycles and 4% grade 4). One episode of grade 4 thrombocytopenia (4%) occurred. Fifty-two percent of the patients had anemia that required transfusions. Eight of 23 patients with measurable disease (35%; 95% confidence interval [CI], 15% to 54%) had partial responses (PRs) lasting longer than 1 month. Two of these patients had minor residual computed tomographic changes but had clinical complete remissions that lasted up to 53 weeks while they were not undergoing further therapy. One patient with nonmeasurable disease had a PR (by CA-125 criteria) that lasted 6 months, for an overall response rate of 38% in nine of 24 patients (95% CI, 18% to 57%). The median time to progression was 26 weeks. Pharmacodynamic analysis demonstrated a statistically significant decrease in free PBMC topo-1 level at weeks 2 and 3 of drug administration. There was a strong statistical correlation between the decrease in free topo-1 levels and increasing area under the curve (AUC) for topotecan. This was confirmed in a pharmacodynamic model. CONCLUSION: Twenty-one-day infusion is a well-tolerated method of administering topotecan. Pharmacodynamic studies demonstrate correlations between (1) the week of infusion and the PBMC topo-1 level, (2) the AUC of topotecan and the decrease in topo-1 levels, and (3) the change in topo-1 level and the neutrophil nadir. The objective response rate of 35% to 38% (95% CI, 15% to 57%) in this small multicenter study is at the upper level for topotecan therapy in previously treated ovarian cancer. Prolonged topotecan administration therefore warrants further investigation in larger, randomized studies comparing this 21-day schedule with the once-daily-for-5-days schedule.


Subject(s)
Antineoplastic Agents/therapeutic use , Ovarian Neoplasms/drug therapy , Topotecan/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Area Under Curve , Cisplatin/therapeutic use , Female , Humans , Infusions, Intravenous , Middle Aged , Ovarian Neoplasms/pathology , Topotecan/administration & dosage , Topotecan/adverse effects , Topotecan/pharmacology , Treatment Outcome
15.
J Clin Oncol ; 17(3): 880-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10071279

ABSTRACT

PURPOSE: To determine the maximum-tolerable dose (MTD) of paclitaxel given as a 3-hour intravenous (IV) infusion that could be used in conjunction with doxorubicin and dexrazoxane, and to determine the effect of dexrazoxane on the pharmacokinetics of paclitaxel and doxorubicin. PATIENTS AND METHODS: Twenty-five patients with advanced breast cancer received dexrazoxane (600 mg/m2 by IV infusion over 15 minutes), followed 15 minutes later by doxorubicin (60 mg/m2 IV), followed 15 minutes later by paclitaxel (150 or 175 mg/m2 by IV infusion over 3 hours) in cohorts of three to six patients using a standard phase I design without (group A) and with (group B) granulocyte colony-stimulating factor (G-CSF). Treatment continued until there was a substantial decrease in the left ventricular ejection fraction (LVEF), congestive heart failure, progressive disease, or physician discretion to discontinue. RESULTS: The MTD of paclitaxel was 150 mg/m2, and adjunctive therapy with G-CSF was required to prevent febrile neutropenia. Dexrazoxane had no significant effect on the pharmacokinetics of paclitaxel or doxorubicin. After a median cumulative doxorubicin dose of 360 mg/m2 (range, 60 to 870 mg/m2), no patient developed congestive heart failure or had a decrease in LVEF below normal. An objective response occurred in all five patients with locally advanced breast cancer and in eight of 20 patients (40%; 95% confidence interval, 19% to 61%) with metastatic breast cancer. CONCLUSION: When combined with doxorubicin (60 mg/m2) and dexrazoxane (600 mg/m2), paclitaxel given as a 3-hour infusion had an MTD of 150 mg/m2, and G-CSF was required to prevent febrile neutropenia. Dexrazoxane had no effect on the pharmacokinetics of paclitaxel or doxorubicin. No patient in this trial had a decrease in the LVEF below normal, compared with about 20% to 50% of patients treated with doxorubicin and paclitaxel without dexrazoxane in other trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Area Under Curve , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Half-Life , Humans , Middle Aged , Neutropenia/chemically induced , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Razoxane/administration & dosage , Razoxane/pharmacology , Remission Induction , Ventricular Function, Left/drug effects
16.
Ann Intern Med ; 130(6): 510-4, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10075619

ABSTRACT

BACKGROUND: Hypericin, the active compound in St. John's Wort, has antiretroviral activity in vitro. Many HIV-infected persons use St. John's wort. OBJECTIVE: To evaluate the safety and antiretroviral activity of hypericin in HIV-infected patients. DESIGN: Phase I study. SETTING: Four clinical research units. PATIENTS: 30 HIV-infected patients with CD4 counts less than 350 cells/mm3. INTERVENTION: Intravenous hypericin, 0.25 or 0.5 mg/kg of body weight twice weekly or 0.25 mg/kg three times weekly, or oral hypericin, 0.5 mg/kg daily. MEASUREMENTS: Safety was assessed at weekly visits. Antiretroviral activity was assessed by changes in HIV p24 antigen level, HIV titer, HIV RNA copies, and CD4 cell counts. RESULTS: Of the 30 patients who were enrolled, 16 discontinued treatment early because of toxic effects. Severe cutaneous phototoxicity was observed in 11 of 23 (48% [95% CI, 27% to 69%]) evaluable patients, and dose escalation could not be completed. Virologic markers and CD4 cell count did not significantly change. CONCLUSIONS: Hypericin caused significant phototoxicity and had no antiretroviral activity in the limited number of patients studied.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Perylene/analogs & derivatives , Administration, Oral , Adult , Anthracenes , Anti-HIV Agents/adverse effects , CD4 Lymphocyte Count , Dermatitis, Phototoxic/etiology , Female , Follow-Up Studies , HIV Core Protein p24/blood , HIV Infections/immunology , HIV Infections/virology , Humans , Injections, Intravenous , Male , Perylene/adverse effects , Perylene/therapeutic use , RNA, Viral/blood , Statistics, Nonparametric , Viral Load
17.
Drug Metab Dispos ; 27(1): 13-20, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9884304

ABSTRACT

Naturally occurring phenethyl isothiocyanate (PEITC) and its synthetic homolog 6-phenylhexyl isothiocyanate (PHITC) are both effective inhibitors of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced lung tumor development in A/J mice and F344 rats. To help explain why PHITC is considerably more efficacious than PEITC in chemopreventive potency, comparative disposition and pharmacokinetics data for male F344 rats were obtained after a single gavage dose of 50 micromol/kg (3.71 microCi/micromol) [14C]PEITC or 50 micromol/kg (6.59 microCi/micromol) [14C]PHITC in corn oil. After [14C]PEITC dosing, whole blood 14C peaked at 2.9 h, with an elimination half-life (T1/2e) of 21.7 h; blood 14C from [14C]PHITC-treated rats peaked at 8.9 h, with an T1/2e of 20.5 h. In lungs, the target organ, the T1/2e for [14C]PHITC and its labeled metabolites were more than twice that for [14C]PEITC and its labeled metabolites. The effective dose (area under the concentration-time curve) for 14C from PHITC was greater than 2.5 times the area under the concentration-time curve of 14C from PEITC in liver, lungs, and several other tissues. During 48 h, approximately 16.5% of the administered dose of [14C]PHITC was expired as [14C]CO2, more than 100 times the [14C]CO2 expired by rats treated with [14C]PEITC. In rats given [14C]PEITC, 88.7 +/- 2.2% and 9.9 +/- 1.9% of the dose appeared in the urine and feces, respectively, during 48 h; however, rats given [14C]PHITC excreted 7.2 +/- 0.8% of the dose of 14C in urine and 47.4 +/- 14.0% in the feces. Higher effective doses of PHITC in the lungs and other organs may be the basis, in part, for its greater potency as a chemopreventive agent.


Subject(s)
Anticarcinogenic Agents/pharmacokinetics , Isothiocyanates/pharmacokinetics , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/blood , Area Under Curve , Biological Availability , Carbon Radioisotopes , Half-Life , Isothiocyanates/administration & dosage , Isothiocyanates/blood , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Rats , Rats, Inbred F344 , Tissue Distribution
18.
J Immunother ; 21(5): 371-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9789199

ABSTRACT

Interleukin 1 alpha (IL-1 alpha) is a cytokine with pleiotropic effects, including cytotoxic-cytostatic activity against some tumor cell lines. We have conducted a phase I study of recombinant human IL-1 alpha (rhIL-1 alpha) in 17 patients with refractory malignancies to examine its toxicity and biologic activity. rhIL-1 alpha was given as a 2-h IV infusion daily for 5 days at five dose levels (0.08, 0.2, 0.8, 2.0, and 5.0 micrograms/m2). Seventeen patients with malignancies were treated, with no objective tumor responses noted. Common toxicities included: fever (100%), rigors and/or chills (96%), myalgia (54%), and headache (48%). Three patients developed grade III hypotension. The maximum tolerated dose was 2.0 micrograms/m2. rhIL-1 alpha induced a significant increase in absolute neutrophil count over baseline (p < 0.05), a delayed but significant increase in platelet count over baseline (p < 0.05), and there was a marked increase in the number of progenitors [colony-forming units (CFU)-G, CFU-M, CFU-GM, CFU-GEMM and burst-forming units (BFU-E)] observed in the peripheral blood. Nine of 12 evaluable patients showed an increase in bone marrow cellularity or myeloid:erthyroid ratio. Immunophenotyping did not demonstrate an increase in peripheral blood or bone marrow CD34+ cells. Interferon-gamma-mediated monocyte cytotoxicity (MCCTX) was significantly enhanced from baseline (p < 0.001), although an increase in direct MCCTX did not reach statistical significance. In summary, rhIL-1 alpha administration is well tolerated at a dose of 2.0 micrograms/m2 with fever, rigors, myalgia, and headache being the most frequent toxicities. Although there were no objective tumor responses, we have demonstrated significant biologic activity with increased neutrophil and platelet counts, increased peripheral blood progenitor cells, and enhanced interferon-gamma-mediated MCCTX.


Subject(s)
Interleukin-1/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Bone Marrow/drug effects , Cytokines/analysis , Cytotoxicity, Immunologic/drug effects , Female , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Infusions, Intravenous , Interleukin-1/administration & dosage , Interleukin-1/adverse effects , Lymphocyte Subsets/drug effects , Male , Middle Aged , Monocytes/drug effects , Neoplasms/immunology , Recombinant Proteins , Safety
19.
Clin Cancer Res ; 4(7): 1679-88, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9676842

ABSTRACT

To evaluate radiometal-labeled humanized BrE-3 (huBrE-3) monoclonal antibody as a radioimmunolocalization and therapeutic agent in breast cancer patients, tumor localization, pharmacokinetics, radiation dosimetry, and immunogenicity of (111)In-labeled combined 1-p-isothiocyanatobenzyl 3-methyl- and 1-p-isothiocyanatobenzyl 4-methyldiethylenetriamine pentaacetic acid (MX-DTPA) huBrE-3 were studied. Seven women with BrE-3 antigen-positive, metastatic breast carcinoma underwent (111)In huBrE-3 infusion (5 mCi; 50 mg), followed by serial gamma camera imaging and plasma sampling. Region of interest analysis of images was used to make radiation absorbed dose estimates for (111)In huBrE-3. Data were extrapolated to 90Y huBrE-3. Human anti-human antibody (HAHA) response was measured in serum samples obtained up to 3 months after infusion. Patients tolerated infusions well. Seventy-six percent of 105 known sites of disease were identified on planar and single-photon emission computed tomography scans. For six of seven patients, a biexponential model fit the plasma time-activity curve best with an average T1/2alpha=10.6+/-8.5 (SD) h and average T1/2beta=114.2+/-39.2 h. Radiation absorbed dose estimates for (111)In huBrE-3 for whole body averaged 0.53+/-.08 rads/mCi. Dose estimates for 90Y huBrE-3 for marrow averaged 8.4+/-11.9 rads/mCi, and for tumors, 70+/-31.5 rads/mCi. Liver radioactivity uptake averaged 19.7+/-8.8% injected dose at 24 h after infusion, translating into an average radiation absorbed dose 21.1+/-12 rads/90Y mCi administered. Only one of seven patients demonstrated a low level of HAHA response. Although the plasma half-lives are longer and marrow dose higher for radiolabeled huBrE-3 compared with the murine construct, the excellent tumor localization, good tumor dosimetry, and low immunogenicity support the use of 90Y-huBrE-3 antibody for radioimmunotherapy of breast cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/radiotherapy , Indium Radioisotopes/therapeutic use , Pentetic Acid/analogs & derivatives , Radioimmunotherapy/methods , Yttrium Radioisotopes/therapeutic use , Adult , Antibodies, Monoclonal/pharmacokinetics , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Female , Humans , Indium Radioisotopes/pharmacokinetics , Middle Aged , Pentetic Acid/pharmacokinetics , Pentetic Acid/therapeutic use , Radiotherapy Dosage , Tomography, Emission-Computed, Single-Photon , Yttrium Radioisotopes/pharmacokinetics
20.
Clin Cancer Res ; 4(3): 545-57, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9533521

ABSTRACT

Analogues of camptothecins are specific inhibitors of eukaryotic DNA topoisomerase I (topo I) that lead to DNA damage and, eventually, cellular cytotoxicity. Camptothecin analogues bind to this target enzyme in the course of its normal function and stabilize the DNA-enzyme adduct to form a "cleavable complex." Preclinical experiments using Western blot analyses have shown cleavable complex formation to be the key intermediate step in topo I inhibition. In this series of experiments, it was our goal to convert this laboratory technique into a useful clinical assay, allowing measurement of the target enzyme and detection of the key intermediate in clinical specimens taken from patients being treated with the topo I inhibitor topotecan. Because available antibodies were not sufficiently sensitive at the start of this project, we identified a highly specific human SCL-70 antibody from a patient with scleroderma, which allowed quantitative determination of topo I copy number in HeLa and HT-29 cell lines. Additional refinements of the Western blot technique were accomplished to improve signal:noise ratio. In surgical tumor specimens, we found the median topo I level to be 30.1 x 10(5) copies/cell for gastric adenocarcinomas, compared to 18.4 x 10(5) copies/cell for normal gastric mucosae in the same samples. For lung adenocarcinoma, the median protein level was 21.5 x 10(5) copies/cell, compared with the normal tissue counterpart protein level of 12.7 x 10(5) copies/cell. The median tumor:normal ratios from paired samples of these tumor types were 1.51 and 1.84, respectively. As part of a Phase II study evaluating the efficacy of topotecan (1.5-2.0 mg/m2 daily for 5 days) in upper gastrointestinal malignancies, we obtained tumor and normal mucosa biopsies in 11 patients with gastric or esophageal cancer, 30 min after administration on day 4 or 5. Three patients with gastric adenocarcinoma had stable disease as their best response, with the remainder of patients progressing. Improvement in Western blotting methodology allowed the quantitation of topo I levels in these gastric and esophageal cancer biopsies, which could be augmented by brief heating to release complexed topo I. We were also able to directly visualize high molecular weight topo I-containing bands, which were shown to be cleavable complexes by heat reversal, with restoration of the topo I Mr 100,000 band. Using this heat reversal technique, we determined the presence of cleavable complex in a total of 7 of 11 patient biopsy samples (5 tumors and 2 normal mucosae). In patients treated with topotecan on this dose and schedule, we determined that a median of 73% of the total tumor topo I was involved in cleavable complex (range, 18.3-91%). The intensity of the Mr 100,000 topo I band in biopsy specimens of patients receiving topotecan represented "free" or noncomplexed topo I. The median copy number for the residual, noncomplexed topo I (n = 11) was 7.36 x 10(5) copies/cell, significantly less than the median of 30.1 x 10(5) copies/cell for random tumor specimens from patients with gastric adenocarcinomas (P < 0.001). Pharmacodynamic analysis demonstrated a negative correlation between the noncomplexed topo I copy number and topotecan area under the curve (Spearman rank test: r(s) = -0.81, P = 0.003). Nonlinear regression analyses of these data were best fit with an inhibitory maximum effect model, yielding parameter estimates for Emax and EC50 of 29.3 x 10(5) copies/cell (coefficient of variation = 22%) and 43.1 ng x h/ml (coefficient of variation = 27%), respectively. Through a series of careful modifications and refinements, we have improved the Western blot assay for topo I for use in clinical monitoring. We have demonstrated the ability to directly visualize cleavable complex in patients being treated with topo I inhibitor therapy and have directly quantitated free topo I, as well as the key topo I intermediate (cleavable complex), in biopsy specimens obtained from pat


Subject(s)
Antineoplastic Agents/therapeutic use , DNA Topoisomerases, Type I/metabolism , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/enzymology , Topotecan/therapeutic use , Adenoma, Islet Cell/enzymology , Antineoplastic Agents/blood , Autoantibodies/blood , Autoantigens/immunology , Biomarkers/blood , Biopsy , Breast Neoplasms/enzymology , Cell Line , Endometrial Neoplasms/enzymology , Esophageal Neoplasms/enzymology , Female , Gastrointestinal Neoplasms/pathology , HeLa Cells , Humans , Lung Neoplasms/enzymology , Nuclear Proteins/immunology , Ovarian Neoplasms/enzymology , Pancreatic Neoplasms/enzymology , Scleroderma, Systemic/blood , Scleroderma, Systemic/immunology , Stomach Neoplasms/enzymology , Topoisomerase I Inhibitors , Topotecan/blood , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...