Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Ann Oncol ; 29(5): 1304-1311, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29788155

ABSTRACT

BACKGROUND: Checkpoint kinase 1 (Chk1) inhibition following chemotherapy-elicited DNA damage overrides cell cycle arrest and induces mitotic catastrophe and cell death. GDC-0575 is a highly-selective oral small-molecule Chk1 inhibitor that results in tumor shrinkage and growth delay in xenograft models. We evaluated the safety, tolerability, and pharmacokinetic properties of GDC-0575 alone and in combination with gemcitabine. Antitumor activity and Chk1 pathway modulation were assessed. PATIENTS AND METHODS: In this phase I open-label study, in the dose escalation stage, patients were enrolled in a GDC-0575 monotherapy Arm (1) or GDC-0575 combination with gemcitabine Arm (2) to determine the maximum tolerated dose. Patients in arm 2 received either i.v. gemcitabine 1000 mg/m2 (arm 2a) or 500 mg/m2 (arm 2b), followed by GDC-0575 (45 or 80 mg, respectively, as RP2D). Stage II enrolled disease-specific cohorts. RESULTS: Of 102 patients treated, 70% were female, the median age was 59 years (range 27-85), and 47% were Eastern Cooperative Oncology Group PS 0. The most common tumor type was breast (37%). The most frequent adverse events (all grades) related to GDC-0575 and/or gemcitabine were neutropenia (68%), anemia (48%), nausea (43%), fatigue (42%), and thrombocytopenia (35%). Maximum concentrations of GDC-0575 were achieved within 2 hours of dosing, and half-life was ∼23 hours. No pharmacokinetic drug-drug interaction was observed between GDC-0575 and gemcitabine. Among patients treated with GDC-0575 and gemcitabine, there were four confirmed partial responses, three occurring in patients with tumors harboring TP53 mutation. Pharmacodynamic data were consistent with GDC-0575 inhibition of gemcitabine-induced expression of pCDK1/2. CONCLUSION: GDC-0575 can be safely administered as a monotherapy and in combination with gemcitabine; however, overall tolerability with gemcitabine was modest. Hematological toxicities were frequent but manageable. Preliminary antitumor activity was observed but limited to a small number of patients with a variety of refractory solid tumors treated with GDC-0575 and gemcitabine. CLINICAL TRIAL NUMBER: NCT01564251.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Pyrroles/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Checkpoint Kinase 1/antagonists & inhibitors , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Fatigue , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea , Neutropenia/chemically induced , Neutropenia/epidemiology , Piperidines/adverse effects , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Thrombocytopenia , Treatment Outcome , Gemcitabine
2.
Br J Cancer ; 117(11): 1592-1599, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28972963

ABSTRACT

BACKGROUND: ARQ 087 is an orally administered pan-FGFR inhibitor with multi-kinase activity. This Phase 1 study evaluated safety, pharmacokinetics, and pharmacodynamics of ARQ 087 and defined the recommended Phase 2 dose (RP2D). METHODS: Patients with advanced solid tumours received ARQ 087 administered initially at 25 mg every other day and dose-escalated from 25 to 425 mg daily (QD) continuous dosing. FGF19, 21, 23, and serum phosphate were assessed as potential biomarkers of target engagement. RESULTS: 80 patients were enrolled, 61 in dose-escalation/food-effect cohorts and 19 with pre-defined tumour types in the expansion cohort. The most common ARQ 087-related adverse events were fatigue (49%), nausea (46%), aspartate aminotransferase (AST) increase (30%), and diarrhoea (23%). Four patients (5%) experienced grade 1 treatment-related hyperphosphataemia. Dose-limiting toxicity was reversible grade 3 AST increase. The RP2D was 300 mg QD. Pharmacokinetics were linear and dose-proportional from 25 to 325 mg QD, and were unaffected by food. Statistically significant changes (P-value<0.05) suggest phosphate and FGF19 levels as markers of target engagement. In 18 evaluable patients with FGFR genetic alterations, 3 confirmed partial responses (two intrahepatic cholangiocarcinomas (iCCA) with FGFR2 fusions and one urothelial cancer with FGFR2 and FGF19 amplification) and two durable stable disease at ⩾16 weeks with tumour reduction (FGFR2 fusion-positive iCCA and adrenocortical carcinoma with FGFR1 amplification) were observed. CONCLUSIONS: ARQ 087 had manageable toxicity at the RP2D of 300 mg QD, showed pharmacodynamics effects, and achieved objective responses, notably in patients with FGFR2 genetic alterations.


Subject(s)
Aniline Compounds/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aniline Compounds/adverse effects , Aniline Compounds/pharmacokinetics , Female , Humans , Male , Middle Aged , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Receptors, Fibroblast Growth Factor/genetics
3.
Ann Oncol ; 28(8): 1776-1787, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28838216

ABSTRACT

Epigenetics has been defined as 'the structural adaptation of chromosomal regions so as to register, signal or perpetuate altered activity states.' Currently, several classes of anticancer drugs function at the epigenetic level, including inhibitors of DNA methyltransferase, histone deacetylase (HDAC), lysine-specific demethylase 1, zeste homolog 2, and bromodomain and extra-terminal motif (BET) proteins.BET proteins have multiple functions, including the initiation and elongation of transcription and cell cycle regulation. In recent years, inhibitors of BET proteins have been developed as anticancer agents. These inhibitors exhibit selectivity for tumor cells by preferentially binding to superenhancers, noncoding regions of DNA critical for the transcription of genes that determine a cell's identity. Preclinical research on BET inhibitors has identified them as a potential means of targeting MYC.Early clinical trials with BET inhibitors have had mixed results, with few responses in both hematologic and solid tumors that tend to be short-lived. Toxicities have included severe, thrombocytopenia, fatigue, nausea, vomiting, and diarrhea; GI side-effects, fatigue, and low-grade dysgeusia have limited compliance. However, preclinical data suggest that BET inhibitors may have a promising future in combination with other agents. They appear to be able to overcome resistance to targeted agents and have strong synergy with immune checkpoint inhibitors as well as with multiple epigenetic agents, particularly HDAC inhibitors. In many instances, BET and HDAC inhibitors were synergistic at reduced doses, suggesting a potential means of avoiding the overlapping toxicities of the two drug classes.BET inhibitors provide a novel approach to epigenetic anticancer therapy. However, to date they appear to have limited efficacy as single agents. A focus on BET inhibitors in combination with other drugs such as targeted and/or as other epigenetic agents is warranted, due to limited monotherapy activity, including pharmacodynamic correlatives differential activity amongst select drug combinations.


Subject(s)
Epigenesis, Genetic , Proteins/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Drug Synergism , Genes, myc , Humans , Neoplasms/drug therapy , Neoplasms/genetics
4.
Ann Oncol ; 27(3): 526-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26646757

ABSTRACT

BACKGROUND: Fibroblast growth factors (FGFs) play important roles in multiple cancers by supporting tumor growth and angiogenesis. FP-1039 (GSK3052230) is a FGF ligand trap consisting of the extracellular domain of FGF receptor 1 (FGFR1) fused with the Fc region of IgG1. FP-1039 binds and neutralizes multiple FGFs that normally bind FGFR1. The primary objective of this phase I study was to evaluate the safety and tolerability of FP-1039. PATIENTS AND METHODS: Eligible patients with metastatic or locally advanced solid tumors for which standard treatments were ineffective were treated with weekly doses of FP-1039 for 4 weeks, followed by 2 weeks observation. RESULTS: Thirty-nine subjects received a mean of 6 infusions of FP-1039 at doses ranging from 0.5 to 16 mg/kg weekly, with no maximally tolerated dose identified. Grade 3 or greater treatment emergent adverse events were uncommon. Four dose-limiting toxicities were reported at doses of 0.75 mg/kg (urticaria), 1 mg/kg (intestinal perforation and neutropenia), and 16 mg/kg (muscular weakness). Drug exposure was dose proportional, and the terminal elimination half-life was 2.6-3.9 days following a single dose. Target engagement as measured by low free plasma FGF2 levels was achieved. FGF pathway dysregulation was uncommon. No objective responses were observed. CONCLUSION: In nonselected cancer patients with advanced disease, treatment with FP-1039 was well tolerated and toxicities associated with small molecule drugs that inhibit FGFR tyrosine kinases, including hyperphosphatemia, were not observed. Further studies of FP-1039 in patients selected for FGF pathway dysregulation, who are most likely to benefit, are now underway.


Subject(s)
Antineoplastic Agents/therapeutic use , Fibroblast Growth Factors/antagonists & inhibitors , Immunoglobulin G/therapeutic use , Neoplasms/drug therapy , Oncogene Proteins, Fusion/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Fibroblast Growth Factors/metabolism , Humans , Immunoglobulin G/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Oncogene Proteins, Fusion/adverse effects , Oncogene Proteins, Fusion/pharmacokinetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Fusion Proteins
5.
Br J Cancer ; 104(12): 1862-8, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21610706

ABSTRACT

BACKGROUND: This phase I, open-label, first-in-human study determined dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) of PD 0332991, an oral cyclin-dependent kinase 4/6 inhibitor with potent anti-proliferative activity in vitro/vivo. METHODS: A total of 33 patients with retinoblastoma protein-positive advanced solid tumours or non-Hodgkin's lymphoma refractory to standard therapy or for which no therapy was available received PD 0332991 once daily (QD) for 14 days followed by 7 days off treatment (21-day cycles; Schedule 2/1). RESULTS: Six patients had DLTs (18%; four receiving 200 mg QD; two receiving 225 mg QD); the MTD was 200 mg QD. Treatment-related, non-haematological adverse events occurred in 29 patients (88%) during cycle 1 and 27 patients (82%) thereafter. Adverse events were generally mild-moderate. Of 31 evaluable patients, one with testicular cancer achieved a partial response; nine had stable disease (≥10 cycles in three cases). PD 0332991 was slowly absorbed (mean T(max) 4.2 h) and eliminated (mean half-life 26.7 h). Volume of distribution was large (mean 3241 l) with dose-proportional exposure. Using a maximum effective concentration model, neutropenia was proportional to exposure. CONCLUSION: PD 0332991 was generally well tolerated, with DLTs related mainly to myelosuppression. The MTD, 200 mg QD, is recommended for phase II study.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Piperazines/administration & dosage , Pyridines/administration & dosage , Adult , Aged , Drug Administration Schedule , Female , Humans , Lymphoma, Non-Hodgkin/drug therapy , Male , Maximum Tolerated Dose , Middle Aged , Piperazines/adverse effects , Piperazines/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics , Retinoblastoma Protein/analysis
6.
J Cell Biochem ; 99(4): 1021-39, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16927308

ABSTRACT

It has been well established that a functioning vascular supply is essential for solid tumor growth and metastases. In the absence of a viable vascular network, tumors are unable to grow beyond a few millimeters and therefore remain dormant. Initiation of angiogenesis allows for continued tumor growth and progression. Targeting tumor vasculature, either by inhibiting growth of new tumor blood vessels (antiangiogenic agents) or by destroying the already present tumor vessels (vascular disrupting agents; VDA), is an area of extensive research in the development of new antitumor agents. These two groups differ in their direct physiological target, the type or extent of disease that is likely to be susceptible, and the treatment schedule. VDAs target the established tumor blood vessels, resulting in tumor ischemia and necrosis. These agents show more immediate effects compared to antiangiogenic agents and may have more efficacy against advanced bulky disease. VDAs can be divided into two groups--ligand-bound and small molecule agents. Both VDA groups have demonstrated antitumor effects and tumor core necrosis, but consistently leave a thin rim of viable tumor cells at the periphery of the tumor. More evidence suggests VDAs will have their greatest effect in combination with conventional chemotherapy or other modes of treatment that attack this outer rim of cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/blood supply , Animals , Flavonoids/pharmacology , Ligands , Tubulin/metabolism , Xanthones/pharmacology
7.
Br J Cancer ; 91(3): 418-24, 2004 Aug 02.
Article in English | MEDLINE | ID: mdl-15238978

ABSTRACT

The epidermal growth factor receptor (EGFR) is a member of the erbB family of tyrosine kinase receptors (RTK). The EGFR is involved in cell proliferation, metastasis and angiogenesis, and is expressed in a large proportion of epithelial tumours. The two main classes of EGFR inhibitors in clinical trials are the RTK inhibitors and the monoclonal antibodies. The clinical development of EGFR inhibitors has introduced new challenges to the design of phase I, II, and III trials. Both classes of agents can be safely administered at doses sufficient to inhibit the EGFR system. Receptor tyrosine kinase inhibitors have been extensively evaluated in non-small-cell lung cancer. In this setting, gefitinib has demonstrated activity in patients who fail initial chemotherapy. Monoclonal antibodies have been developed in combination with cytotoxic chemotherapy in several tumour types, most notably colorectal and head and neck cancer. The preliminary results suggest an increase in response rate and time to progression with the combination of cetuximab and chemotherapy in both disease models. Future issues in the development of EGFR inhibitors include the identification of biologic predictors of response, combination with other targeted agents, and their utilisation in earlier stage malignancies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cetuximab , Clinical Trials as Topic , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gefitinib , Humans , Neoplasms/drug therapy , Neoplasms/physiopathology
8.
Invest New Drugs ; 21(1): 63-74, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12795531

ABSTRACT

The spicamycin derivative KRN5500 was considered as a potential anti-cancer agent based on in vitro and preclinical studies. A Phase I study involving 24 cancer patients in whom tumors were refractory to all other conventional therapies was conducted to determine the dose limiting toxicity, maximum tolerated dose, effectiveness, and pharmacokinetic parameters of this drug administered by 1-h IV infusion daily for five consecutive days every 3 weeks. Using an accelerated dose titration strategy, 8.4 mg/m2/d x 5 days was the maximum administered dose. Severe gastrointestinal and hepatic toxicities were observed at doses at or above 4.3 mg/m2/d x 5. The recommended Phase II dose i s 4.3mg/m2/d x 5. The distribution of KRN5500 followed a two-compartment model, and clearance did not decrease significantly over the dose range 0.8-8.4 mg/m2/d x 5. No significant correlation was observed between plasma levels and toxicity. No tumor responses were observed among the 14 patients evaluable for response.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Purine Nucleosides/administration & dosage , Purine Nucleosides/pharmacokinetics , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Purine Nucleosides/adverse effects , Purine Nucleosides/pharmacology , Time Factors , Treatment Outcome
9.
Clin Cancer Res ; 7(11): 3336-42, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11705845

ABSTRACT

PURPOSE: XK469 (2-[4-(7-chloro-2-quinoxalinyloxy) phenoxy]propionic acid), a synthetic quinoxaline phenoxypropionic acid derivative, has broad activity against murine tumors and is entering Phase I clinical development as a topoisomerase IIbeta inhibitor. This study investigated the underlying molecular mechanism of XK469's effects on the cell cycle. EXPERIMENTAL DESIGN: Growth inhibition, cell cycle arrest, induction of p53 and p21 mRNA and protein, and cdc2 phosphorylation and kinase activity were studied in treated cells from the H460 lung cancer line and p21 and p53 knockout cells of the HCT 116 colon cancer line. RESULTS: XK469 arrested H460 cells at G(2)-M, which was associated with cdc2 phosphorylation and decreased cdc2 kinase activity. Moreover, XK469 stabilized p53 and subsequently increased p21(WAF1/CIP1). Furthermore, HCT116 p21(-/-) cells were less sensitive than wild-type cells to XK469-induced growth inhibition, but p53(+/+) and p53(-/-) cells were equally sensitive despite the absence of p21 induction in the p53(-/-) cells. CONCLUSIONS: When considered with published data, our study suggests a complex mechanism of XK469-mediated anticancer activity involving multiple pathways, including p53-dependent and -independent G(2)-M arrest via inactivation of cdc2-cyclin B1 kinase activity.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Quinoxalines/pharmacology , Tumor Suppressor Protein p53/physiology , Cell Cycle/physiology , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/physiology , Dose-Response Relationship, Drug , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitosis/drug effects , Mutation , Neoplasms/pathology , Neoplasms/prevention & control , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
10.
Invest New Drugs ; 19(1): 1-11, 2001.
Article in English | MEDLINE | ID: mdl-11291827

ABSTRACT

OBJECTIVES: CI-994 (N-acetyl dinaline, PD 123654) is a novel oral agent active in a broad variety of murine and human tumor xenografts. While cytotoxic in the Brown Norway (BN) rat leukemia model, growth inhibition in other murine and human tumor xenografts is predominantly cytostatic. Its specific mechanism of action remains unknown. Following CI-994 administration, inhibition of both histone deacetylation and cellular proliferation at the G1 to S transition phase of the cell cycle are observed. This Phase 1 study in patients with solid tumors was carried out to determine a maximum tolerated daily oral dose (MTD) for CI-994 administered on a chronic basis. METHODS: Fifty-three patients received CI-994 daily for treatment durations ranging from 2 to 10 weeks. Dosage escalation proceeded in 2 phases; an Acute Dosing Phase (n = 11) to define the MTD for CI-994 administered over 2 weeks and a Chronic Dosing Phase (n = 29) to define the MTD for daily administration for 8 weeks. Upon completion of the Chronic Dosing Phase, a third cohort of patients (n = 13) received CI-994 at the recommended Phase 2 dose and schedule with 2 additional single doses of drug administered separated by a 1-week washout to assess the effect of food on CI-994 pharmacokinetics. RESULTS: Thrombocytopenia was dose limiting at the MTD of 8 mg/m2/day for 8 weeks. Other toxicities included fatigue and gastrointestinal effects such as nausea, vomiting, diarrhea, constipation and mucositis. Pharmacokinetic studies revealed that peak plasma levels and AUC's generally increased with dose and that food intake did not affect the rate or extent of drug absorption. One patient with heavily pre-treated adenocarcinoma of the lung achieved a Partial Response (PR) lasting over 2 years and 3 additional patients achieved Stable Disease (SD), 1 each with non-small cell lung, colorectal, and renal cancer. CONCLUSIONS: The recommended Phase 2 starting dose is 8 mg/m2/day for 8 weeks repeated after a 2-week drug-free interval.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Phenylenediamines/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Benzamides , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Phenylenediamines/adverse effects , Phenylenediamines/pharmacokinetics , Treatment Outcome
11.
Clin Cancer Res ; 6(8): 3088-94, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10955788

ABSTRACT

SR233377 is a novel thioxanthenone analogue that demonstrated solid tumor selectivity in vitro with activity confirmed in vivo against several murine tumors including those of colon, pancreas, and mammary origin. Its primary preclinical dose-limiting toxicities included myelosuppression and neurological toxicity. The neurological toxicity was acute and could be ameliorated in mice when the drug was administered as a 1-h infusion instead of rapid i.v. injection. As a result of its preclinical efficacy profile, SR233377 entered Phase I clinical investigation. The compound was administered i.v. over 2 h on day 1 repeated every 28 days. The starting dose was 33 mg/m2 (one-tenth the mouse LD10). Escalations continued to 445 mg/m2 (six escalations), where dose-limiting toxicity was observed. At this dose, acute ventricular arrhythmias, including one patient with torsades de pointes and transient cardiac arrest, occurred. Because this toxicity might have been related to the plasma peak, the protocol was amended to a 24-h infusion beginning at 225 mg/m2. With this dose, prolongation of the corrected QT interval (QTc) over the pretreatment levels resulted. Because prolonged QTc is a known forerunner to acute ventricular arrhythmias, clinical development of SR233377 was stopped. However, preclinical antitumor and toxicity studies with analogues are underway with hopes of identifying a new clinical candidate with similar antitumor effects that is devoid of cardiac toxic effects.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Neoplasms/metabolism , Sulfonamides/pharmacokinetics , Thioxanthenes/pharmacokinetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Heart Diseases/chemically induced , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/drug therapy , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Thioxanthenes/adverse effects , Thioxanthenes/therapeutic use
12.
Clin Cancer Res ; 6(6): 2474-81, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10873102

ABSTRACT

Dose intensity, defined as dose administered per unit time, has emerged as a potentially important measurement of anticancer drug exposure and determinant of efficacy. There are several strategies for increasing dose intensity, one being a protracted daily dosing strategy without major dose reduction for toxicity. This strategy involves continued therapy during periods of recovery from reversible toxicity, and it inherently challenges our understanding that renewing tissues cannot repopulate (recover) in the continued presence of cytotoxic drug. We have tested this idea directly in a murine preclinical trial. Specifically, we have tested whether acutely myelotoxic doses of gemcitabine (i.p. injection, 6.0 mg/m2/day), acetyldinaline [CI-994; GOE 5549; PD 123 654; 4-acetylamino-N-(2'-aminophenyl)-benzamide, 150 mg/m2/day p.o.], and/or melphalan (i.p. injection, 7.2 mg/m2/day) can be tolerated for 28 consecutive days and whether suppressed bone marrow function recovers despite this protracted daily therapy. The three drugs all caused acute neutropenia and suppression of medullary hematopoiesis. Damage to progenitor populations exposed to acetyldinaline and gemcitabine was not as severe as that caused by melphalan, in which case absolute neutrophil count, mature progenitors (colony-forming unit granulocyte/macrophage), and immature progenitors (colony-forming unit-S) progressively declined to severely depressed levels. Marrow recovery was observed during continued daily treatment with acetyldinaline and gemcitabine but not melphalan, and marrow function completely recovered after finishing the 28-day course. Pharmacology studies proved that protracted therapy causes little, if any, change in cellular drug tolerance or systemic exposure.


Subject(s)
Antineoplastic Agents/administration & dosage , Deoxycytidine/administration & dosage , Melphalan/administration & dosage , Phenylenediamines/administration & dosage , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Benzamides , Blood Cell Count , Blood Platelets/drug effects , Bone Marrow/drug effects , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Female , Melphalan/adverse effects , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neutropenia/chemically induced , Neutrophils/drug effects , Phenylenediamines/adverse effects , Random Allocation , Stem Cells/drug effects , Time Factors , Gemcitabine
13.
Oncology (Williston Park) ; 14(12 Suppl 11): 33-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11204661

ABSTRACT

Oxaliplatin (Eloxatin) has demonstrated significant activity in a variety of tumor types in addition to colorectal cancer. Several studies have reported on the effectiveness of oxaliplatin as single-agent treatment or in combination with cisplatin (Platinol) or paclitaxel (Taxol) in pretreated advanced ovarian cancer patients, with promising data reported for the combination of oxaliplatin and cisplatin as first-line therapy. Other small studies have shown the activity of single-agent oxaliplatin in anthracycline-resistant metastatic breast cancer and refractory non-Hodgkin's lymphoma. Data have also demonstrated the activity of oxaliplatin in combination with gemcitabine (Gemzar) in advanced pancreatic cancer. In recurrent germ-cell cancer, a "biplatin" regimen of oxaliplatin plus cisplatin was found to be effective salvage therapy. Data from these studies indicate that oxaliplatin is active in both platinum-resistant disease and in tumor types that have previously been unresponsive to platinum treatment. Moreover, it enhances the effect of cisplatin or carboplatin, which is a striking demonstration of differing mechanisms of action. Ongoing and planned trials will evaluate the efficacy of oxaliplatin in other disease settings and combinations.


Subject(s)
Neoplasms/drug therapy , Organoplatinum Compounds/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Clinical Trials, Phase II as Topic , Female , Humans , Middle Aged , Oxaliplatin , Paclitaxel/therapeutic use
14.
J Clin Oncol ; 17(2): 485-93, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10080589

ABSTRACT

PURPOSE: Capecitabine is a novel, oral, selectively tumor-activated fluoropyrimidine carbamate. This large multicenter phase II trial tested the efficacy and safety of twice-daily oral capecitabine at 2,510 mg/m2/d given for 2 weeks followed by a 1-week rest period and repeated in 3-week cycles, in patients with paclitaxel-refractory metastatic breast cancer. PATIENTS AND METHODS: Patients were to have received at least two but not more than three prior chemotherapeutic regimens, one of which had to have contained paclitaxel given for metastatic disease. One hundred sixty-three patients were entered onto the study at 25 centers, and 162 patients received capecitabine. One hundred thirty-five patients had bidimensionally measurable disease, and 27 patients had assessable disease. RESULTS: The overall response rate was 20% (95% confidence interval, 14% to 28%). All responding patients were resistant to or had failed paclitaxel, and all had received an anthracycline. Three complete responses were seen, with complete response durations of 106, 109, and 194+ days. Median duration of response was 8.1 months, median survival time was 12.8 months, and the median time to disease progression was 93 days. The most common treatment-related adverse events were hand-foot syndrome, diarrhea, nausea, vomiting, and fatigue. Diarrhea (14%) and hand-foot syndrome (10%) were the only treatment-related adverse events that occurred with grade 3 or 4 intensity in more than 10% of patients. CONCLUSION: Capecitabine is an active drug in the treatment of paclitaxel-refractory metastatic breast cancer. It has a favorable toxicity profile with the added advantage of being an oral drug administered at home.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Capecitabine , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Disease Progression , Drug Resistance, Neoplasm , Female , Fluorouracil/analogs & derivatives , Humans , Middle Aged , Paclitaxel/therapeutic use , Survival Analysis
15.
Cancer Chemother Pharmacol ; 43(2): 162-4, 1999.
Article in English | MEDLINE | ID: mdl-9923823

ABSTRACT

PURPOSE: We completed a phase II trial of CI-958 (NSC 635371) in patients with advanced colorectal cancer given at a dose of 700 mg/m2 every 21 days. METHODS: All 15 patients had metastatic disease and had been previously treated with one 5-fluorouracil-based regimen in an adjuvant (six) or metastatic (nine) setting. RESULTS: None of the patients treated with CI-958 had an objective response to treatment. Median survival was 4.8 months after the start of treatment. Leukopenia was the major toxicity, but no patient experienced febrile neutropenia. An acute febrile reaction was seen after infusion in four of the first nine patients treated. This was abrogated by pretreatment with dexamethasone in the remaining patients. CONCLUSIONS: CI-958 was not effective at this dose and schedule in patients with previously treated advanced colorectal cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Indazoles/therapeutic use , Intercalating Agents/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Drug Administration Schedule , Female , Humans , Indazoles/administration & dosage , Indazoles/adverse effects , Intercalating Agents/administration & dosage , Intercalating Agents/adverse effects , Leukopenia/chemically induced , Male , Middle Aged
16.
Invest New Drugs ; 16(4): 287-96, 1998.
Article in English | MEDLINE | ID: mdl-10426660

ABSTRACT

XK469 (NSC 656889) is a water-soluble member of the novel quinoxaline family of antitumor agents. In vitro, XK469 demonstrated selective cytotoxicity for several murine solid tumors including colorectal and mammary adenocarcinoma cell lines, when compared to both leukemia and normal epithelial cells. In vivo, XK469 was active against 7/7 murine tumors tested, including pancreatic ductal carcinomas #02 and #03, colon adenocarcinomas #38 and #51/A, mammary adenocarcinoma #16/C and the Adriamycin resistant mammary adenocarcinomas #16/C/ADR and #17/ADR. XK469 was efficacious both intravenously and orally. Regardless of dosing schedule, conventional mice tolerated higher total doses than SCID or nu/nu mice did. Despite these reduced doses, XK469 was active against xenografts of 4/6 human tumor lines including mammary adenocarcinoma MX-1, the small cell lung cancer DMS 273, the prostate model LNCaP and the CNS tumor SF295. The lower doses in the xenograft studies were below curative levels. The dose-limiting toxicity appeared to be myelosuppression with rapid host recovery (5-8 days), and in vitro assays of XK469 toxicity to murine bone marrow neutrophil progenitors CFU-GM (colony forming unit-granulocyte/macrophage) demonstrated concentration-dependent toxicity from 0.5-30 microg/mL. The difference in drug tolerance between BDF1 and SCID mice was detected in vitro as a 3-fold difference in the IC90 for CFU-GM, despite similar IC50 values. Comparative in vitro hematotoxicology studies revealed that human bone marrow CFU-GM tolerated XK469 as well as their SCID counterparts (IC90 values 5.7 vs. 7.4 microg/mL). Based on comparison with previously tested anti-cancer agents, these data suggest that humans will be able to tolerate XK469 doses that are efficacious against human tumor xenografts.


Subject(s)
Antineoplastic Agents/pharmacology , Quinoxalines/pharmacology , Animals , Antineoplastic Agents/toxicity , Bone Marrow Cells/drug effects , Cell Division/drug effects , Drug Screening Assays, Antitumor , Humans , Mice , Mice, SCID , Quinoxalines/toxicity , Tumor Cells, Cultured
17.
Invest New Drugs ; 15(3): 187-94, 1997.
Article in English | MEDLINE | ID: mdl-9387041

ABSTRACT

CI-994, a substituted benzamide derivative, is a compound that showed solid tumor selectivity for a variety of solid tumor models compared to L1210 leukemia. Due to its lack of aqueous solubility, it requires oral administration. Female B6D2F1 mice were treated with CI-994 once daily by oral administration of 50 mg/kg for 14 days. Following treatment mice were evaluated for pharmacodynamic effects as well as the pharmacokinetic behavior of CI-994 and the de-acetylated derivative dinaline. Mice samples (plasma, urine, feces) were analyzed using solid phase extraction, reverse phase HPLC and ultraviolet detection. The plasma distribution and elimination half-lives for CI-994 were 51 minutes and 9.4 hours, respectively, on D-1; 31 minutes and 3.4 hours, respectively on D-14. The apparent plasma distribution and elimination half-lives for dinaline were 27 minutes and 2.4 hours, respectively, on D-1; 40 minutes and 7.3 hours, respectively on D-14. The CI-994 AUC on D-1 and D-14 were 2879 and 2407 micrograms/ml x minutes, respectively; while the dinaline AUC on D-1 and D-14 were 87 and 92 micrograms/ml x minutes, respectively. Urinary excretion for CI-994 and dinaline was higher on D-14, while the fecal excretion was the same on both days. The Colon #38 tumor growth in treated mice was reduced to 22% of that observed in the controls by D-19. The levels of all blood cells were reduced in the treated mice when compared to controls and the total WBC was the most affected (median 38%). Recovery to pretreatment levels occurred quickly following treatment cessation. Phase I evaluation of chronic oral administration of CI-994 is currently ongoing.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Phenylenediamines/pharmacokinetics , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Area Under Curve , Benzamides , Blood Cell Count , Bone Marrow/drug effects , Colonic Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Female , Metabolic Clearance Rate , Mice , Phenylenediamines/pharmacology , Phenylenediamines/toxicity
18.
Clin Cancer Res ; 3(11): 2047-53, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9815596

ABSTRACT

Two new thioxanthenones, 183577 and 232759, have rekindled interest in the development of representatives from this class of structures as useful anticancer agents. Although the mechanism of action is unknown, both compounds demonstrated a similar spectrum of solid tumor selectivity. 232759 was selected for clinical development because it showed no hepatotoxicity in preliminary studies, whereas 183577 showed hepatotoxicity but only at the maximum tolerated dose (MTD). The limiting toxicity for the clinical candidate was myelosuppression in preliminary studies. Plasma and tissue drug levels, as well as protein binding, were studied in mice using optimal administration times at the MTD for each drug (for 183577, this was a 4-h infusion at 1350 mg/m2 and for 232759, it was a 5-min injection at 240 mg/m2), as well as at one-half the MTD for the clinical candidate. The drugs were 96-100% bound by plasma proteins. The peak drug concentrations, half-life, and area under the concentration-time curve in plasma for 183577 were 3483 ng/ml, 465 min, and 2018 microgram/ml. min, respectively. The peak drug concentration, half-life, and area under the concentration-time curve in plasma for 232759 were 5257 ng/ml, 44 min, and 276 microgram/ml. min, respectively, at the MTD and 2810 ng/ml, 40 min, and 110 microgram/ml. min at one-half the MTD. In all instances of simultaneous measurements, drug concentrations were equal or higher in tissues than they were in plasma. Unlike the plasma and kidney concentrations of 183577, the liver concentrations did not show a declining trend over the 8-h observation period. Declines in plasma, liver, kidney, and tumor levels of 232759 were detected over the 8-h observation period. The sustained high 183577 concentration in liver is believed to be responsible for its prolonged half-life and hepatotoxicity. Evidence for metabolism of the parent drugs was based on the finding of additional peaks on the high-pressure liquid chromatography tracings. Future studies will focus on identification and antitumor studies of these presumed metabolites in hopes of a better understanding of the solid tumor activity profiles and toxic effects of these compounds.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacokinetics , Colonic Neoplasms/metabolism , Sulfonamides/pharmacokinetics , Thioxanthenes/pharmacokinetics , Adenocarcinoma/blood , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Blood Proteins/metabolism , Colonic Neoplasms/blood , Female , Half-Life , Kidney/metabolism , Leukemia L1210/blood , Leukemia L1210/metabolism , Liver/metabolism , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Sulfonamides/blood , Sulfonamides/therapeutic use , Thioxanthenes/blood , Thioxanthenes/therapeutic use , Tissue Distribution
19.
Cancer ; 77(4): 691-6, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8616761

ABSTRACT

BACKGROUND: Oxidative DNA damage can result from numerous endogenous metabolic processes as well as from exposure to environmental and dietary oxidants. One important type of oxidative DNA damage is the formation of hydroxylated DNA bases. This type of DNA damage may have a role in carcinogenesis. METHODS: We examined the levels of a hydroxylated thymine residue, 5-hydroxy-methyl-2'-deoxyuridine, in DNA obtained from the peripheral blood of breast cancer patients and control women. The isolated DNA was analyzed for levels of 5-hydroxymethyl-2'-deoxyuridine by gas chromatography with mass spectral detection. RESULTS: The levels of this modified base were significantly higher in 25 breast cancer patients compared with 38 controls, with levels of 0.112 +/- 0.046 in the cancer patients versus 0.083 - 0.025 in the controls, given as pg 5-hydroxymethyl-2'-deoxyuridine/ng thymidine, mean +/- standard deviation (P = 0.019). After controlling for various covariates, the adjusted mean levels of oxidative DNA damage were still significantly higher in women with breast cancer relative to controls. CONCLUSIONS: These results indicate that the levels of 5-hydroxymethyl-2'-deoxyuridine in DNA from peripheral nucleated blood may be potentially useful as a marker of breast cancer.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/diagnosis , DNA Damage , DNA/blood , Thymidine/analogs & derivatives , Breast Neoplasms/blood , DNA/chemistry , Female , Humans , Middle Aged , Odds Ratio , Pilot Projects , Postmenopause , Reference Values , Regression Analysis , Risk Factors , Smoking , Statistics, Nonparametric , Thymidine/analysis
20.
Invest New Drugs ; 13(4): 321-6, 1996.
Article in English | MEDLINE | ID: mdl-8824350

ABSTRACT

CC-1065 is a unique alkylating agent that preferentially binds in the minor groove of double-stranded DNA at adenine-thymine-rich sites. Although it has broad antitumor activity in preclinical models its development was discontinued because of deaths observed during preclinical toxicology studies. Adozelesin is a potent synthetic analog that was chosen for clinical development because it had a similar preclinical antitumor spectrum, but did not produce deaths similar to CC-1065 at therapeutic doses. Phase I evaluations using a variety of Adozelesin treatment schedules have been conducted. This report describes our experience using a multiple dose treatment schedule. Endpoints including antitumor response, maximum tolerated dose, dose limiting toxicity as well as other toxicities and the recommended Phase II starting dose were determined. Adozelesin was given as a 10 minute IV infusion for 5 consecutive days every 21 days or upon recovery from toxicity. The dose range evaluated was 6-30 mcg/m2/day. All patients had refractory solid tumors and had received prior cytotoxic treatment. Thirty-three patients (22 men: 11 women) were entered onto the study and 87 courses were initiated. Dose limiting toxicity was cumulative myelosuppression (leucopenia, thrombocytopenia). The maximum tolerated dose was 30 mcg/m2/day. The only other significant toxicity was an anaphylactoid syndrome that occurred in 2 patients. A partial response was observed in a patient with refractory soft tissue sarcoma. The recommended Phase II starting dose of Adozelesin using a 10 minute IV infusion for 5 consecutive days is 25 mcg/m2/day to be repeated every 4-6 weeks to allow recovery from myelotoxicity, based on our experience. Additional Phase I and II studies with Adozelesin are recommended.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Cyclohexanecarboxylic Acids/therapeutic use , Indoles , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents, Alkylating/adverse effects , Benzofurans , Cyclohexanecarboxylic Acids/adverse effects , Cyclohexenes , Dose-Response Relationship, Drug , Drug Administration Schedule , Duocarmycins , Female , Humans , Infusions, Intravenous , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...