Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
J Comput Assist Tomogr ; 25(4): 624-8, 2001.
Article in English | MEDLINE | ID: mdl-11473196

ABSTRACT

As a preliminary study, we measured the necrosis of advanced hepatocellular carcinoma (HCC) by volume histogram after systemic chemotherapy and correlated it with clinical data. Five patients with advanced HCC secondary to chronic hepatitis and cirrhosis underwent pretreatment and posttreatment MR examination on a 1.5 T MR scanner following systemic chemotherapy. MR sequences included dynamic enhanced fast spoiled gradient echo 3D images. Clinical response to chemotherapy, as determined by MR images, was measured as changes of both the total tumor volume and the percent of tumor necrosis by volume histogram algorithm. Four of five patients had clinical improvement. Three of these patients had no or minimal change of tumor volume; however, there was an increase in tumor necrosis in follow-up MR image. One patient of five with no change in tumor necrosis had no response and died at 3 months. Serial MR images showed increased irregular necrosis of advanced HCC after systemic chemotherapy, but stable volume, in patients who responded clinically to systemic chemotherapy.


Subject(s)
Carcinoma, Hepatocellular/diagnostic imaging , Liver Neoplasms/diagnostic imaging , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Female , Gadolinium , Hepatitis C, Chronic/complications , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Magnetic Resonance Imaging/methods , Male , Middle Aged , Necrosis , Radionuclide Imaging
2.
Int J Hematol ; 72(4): 425-35, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11197208

ABSTRACT

Amifostine (Ethyol), the first broad-spectrum cytoprotectant approved in many countries for clinical use, is an analog of cysteamine and was originally developed by the U.S. Walter Reed Army Institute of Research in the 1950s as a radioprotective agent. Studies have shown that amifostine selectively protects normal tissues of various organs from the effects of radiation and multiple cytotoxic chemotherapeutic drugs. Amifostine has demonstrated broad-spectrum cytoprotection against myelotoxicity, nephrotoxicity, xerostomia, and mucositis associated with various chemotherapy and radiation modalities. Amifostine has been evaluated in large comparative clinical trials in patients with advanced ovarian cancer, rectal cancer, and head and neck cancer, and in many phase 2 trials in patients with various neoplastic diseases. These trials have shown that amifostine delivers protection from the cytotoxic effects of cisplatin, cyclophosphamide, and radiation on various organs. Pretreatment with amifostine has also improved salivary gland tolerance of high-dose radioiodine treatment. Recent unique observations include improvement in cytopenia in patients with myelodysplastic syndrome. This review summarizes preclinical and clinical data on amifostine and includes trials that evaluated the drug's chemoprotective and radioprotective effects and other potential uses in clinical oncology.


Subject(s)
Amifostine/therapeutic use , Animals , Antineoplastic Agents/toxicity , Clinical Trials as Topic , Cytoprotection/drug effects , Humans , Neoplasms/drug therapy , Radiation-Protective Agents/therapeutic use
3.
Stroke ; 30(9): 1907-15, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471444

ABSTRACT

BACKGROUND AND PURPOSE: Calcium-channel blockers (CCBs) reduce systolic blood pressure and stroke-related mortality in stroke-prone spontaneously hypertensive rats (SPSHR). Brain ischemia is associated with loss of intracellular antioxidants. Increased formation of oxygen radicals and oxidation of LDL may enhance arterial vasoconstriction by various mechanisms. CCBs that also exert antioxidative properties in vitro may therefore be particularly useful. To investigate such antioxidant effects in vivo, we determined several parameters of LDL oxidation in SPSHR treated with two 1,4-dihydropyridine-type (1,4-DHP) CCBs of different lipophilic properties and compared them with antioxidant-treated and untreated controls. We also tested whether these drugs decrease the formation of oxidation-specific epitopes in arteries. METHODS: Five groups of 9 to 14 SPSHR each (aged 8 weeks) were treated with 80 mg/kg body wt per day nifedipine, 1 mg or 0.3 mg/kg body wt per day lacidipine, vitamin E (100 IU/d), or carrier for 5 weeks. A group of Wistar-Kyoto rats was used as normotensive control. Plasma samples were taken, and LDL was isolated by ultracentrifugation. Then LDL was exposed to oxygen radicals generated by xanthine/xanthine oxidase reaction (2 mmol/L xanthine+100 mU/mL xanthine oxidase), and several parameters of oxidation were determined. The presence of native apolipoprotein B and oxidation-specific epitopes in the carotid and middle cerebral arteries was determined immunocytochemically. RESULTS: 1,4-DHP CCBs completely prevented mortality. Normotensive Wistar-Kyoto rats showed less oxidation than control SPSHR. Plasma lipoperoxide levels were 0.87+/-0.27 micromol/L in control SPSHR, 0.69+/-0.19 and 0.63+/-0.20 micromol/L in the groups treated with 0.3 and 1 mg lacidipine, respectively, and 0.68+/-0.23 micromol/L in nifedipine-treated animals (P<0.05 versus control SPSHR for all values). Both CCBs significantly decreased formation of conjugated dienes and prolonged the lag time in LDL exposed to oxygen radicals. Similarly, lipoperoxides and malondialdehyde were significantly reduced (P<0.05). Reduced relative electrophoretic mobility and increased trinitrobenzenesulfonic acid reactivity of LDL from treated rats (P<0.01) also indicated that fewer lysine residues of apolipoprotein B were oxidatively modified in the presence of 1,4-DHP CCBs. Finally, these drugs reduced the intimal presence of apolipoprotein B and oxidized LDL (oxidation-specific epitopes) in carotid and middle cerebral arteries. CONCLUSIONS: In the SPSHR model, 1,4-DHP CCBs reduce plasma and LDL oxidation and formation of oxidation-specific epitopes and prolong survival independently of blood pressure modifications. Our results support the concept that the in vivo protective effect of these drugs on cerebral ischemia and stroke may in part result from inhibition of oxidative processes.


Subject(s)
Arteries/metabolism , Calcium Channel Blockers/pharmacology , Cerebrovascular Disorders/genetics , Dihydropyridines/pharmacology , Genetic Predisposition to Disease , Lipoproteins, LDL/antagonists & inhibitors , Rats, Inbred SHR/genetics , Animals , Antioxidants/pharmacology , Apolipoproteins B/metabolism , Arteries/drug effects , Cerebrovascular Disorders/mortality , Epitopes/drug effects , Epitopes/metabolism , Immunohistochemistry , Lipoproteins, LDL/blood , Male , Oxidation-Reduction/drug effects , Rats , Rats, Inbred SHR/metabolism , Rats, Inbred WKY , Reference Values , Vitamin E/pharmacology
5.
Semin Oncol ; 26(2 Suppl 7): 3-21, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10348255

ABSTRACT

Administered before cytotoxic chemotherapy or radiation, the aminothiol, amifostine (Ethyol; Alza Pharmaceuticals, Palo Alto, CA/US Bioscience, West Conshohocken, PA), provides broad-spectrum cytoprotection of various normal tissues without attenuating antitumor response. The basis for the selectivity of action resides in the anabolism of amifostine at the normal tissue site by membrane-bound alkaline phosphatase. Dephosphorylation to the free thiol, WR-1065, is followed by rapid uptake into normal tissues. In contrast, uptake into tumor tissue is slow to negligible. Pretreatment with amifostine provides protection of normal tissues from the cytotoxic effects of alkylating agents, organoplatinums, anthracyclines, taxanes, and radiation. Additionally, the mutagenic and carcinogenic effects of these modalities are also attenuated. Preclinical studies show significant protection of marrow progenitor cells. Synergistic effects in marrow recovery are noted with the sequential use of amifostine and granulocyte colony-stimulating factor. Protection of kidneys and neural tissues from cisplatin toxicity has been shown, as well as protection of the heart, intestinal crypt cells, and pulmonary tissues from chemotherapy and radiation, and vasculoconnective and musculoconnective tissue in an irradiated field. Comparative in vitro and in vivo studies using murine and human tumor xenografts show no protection of antitumor effects of these same therapies despite the protection of normal organs. The unique preclinical profile of amifostine serves as the basis for the clinical development program for this new broad-spectrum cytoprotective agent.


Subject(s)
Amifostine/pharmacology , Antineoplastic Agents/adverse effects , Cytoprotection , Prodrugs/pharmacology , Protective Agents/pharmacology , Amifostine/pharmacokinetics , Amifostine/therapeutic use , Animals , Drug Evaluation, Preclinical , Humans , Mercaptoethylamines/pharmacology , Neoplasms/therapy , Neoplasms, Experimental/therapy , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Protective Agents/pharmacokinetics , Protective Agents/therapeutic use , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Tumor Cells, Cultured
6.
Semin Oncol ; 26(2 Suppl 7): 72-81, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10348264

ABSTRACT

Cisplatin, a heavy metal complex, is one of the most active drugs used in the treatment of a variety of cancers. One of the major limitations to the maximization of its therapeutic potential is nephrotoxicity. Several preclinical studies have shown that pretreatment of mice or rats with amifostine (WR-2721, Ethyol; Alza Pharmaceuticals, Palo Alto, CA/US Bioscience, West Conshohocken, PA) protected against nephrotoxicity induced by both single and repeated doses of cisplatin without affecting the antitumor effects of cisplatin. The preclinical evidence of amifostine's protective effects led to phase I-III clinical studies. A phase III trial was conducted in 242 women with stage III/IV ovarian cancer receiving six cycles of cyclophosphamide/cisplatin (CP) +/- amifostine. Consistent with the cumulative nature of cisplatin-induced nephrotoxicity, by cycles 5 and 6, a significantly greater proportion of patients in the control arm compared with patients in the amifostine-treated arm were not eligible to receive cisplatin as scheduled because their serum creatinine levels had failed to return to < or = 1.5 mg/dL. Amifostine pretreatment did not affect the antitumor effects of CP as assessed by response determined at second-look surgery or overall survival. Phase II trials support these findings. To date, amifostine is the only available therapy that can ameliorate the cumulative nephrotoxic effects of cisplatin without reducing antitumor efficacy.


Subject(s)
Amifostine/pharmacology , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Cytoprotection , Kidney/drug effects , Protective Agents/pharmacology , Amifostine/administration & dosage , Amifostine/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/administration & dosage , Clinical Trials as Topic , Cyclophosphamide/administration & dosage , Drug Evaluation, Preclinical , Female , Humans , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Mice , Ovarian Neoplasms/drug therapy , Protective Agents/administration & dosage , Protective Agents/therapeutic use , Rats
7.
Oncology (Williston Park) ; 13(1): 47-59; discussion 63, 67, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10027198

ABSTRACT

Amifostine (Ethyol) is an analog of cysteamine that selectively protects normal tissues in multiple organ systems against the toxic effects of radiation and various cytotoxic drugs while preserving the antitumor effects of these therapies. Amifostine was evaluated in a multicenter, multinational phase III clinical trial that enrolled women with stage III/IV ovarian cancer. Its effects have also been studied using normal human bone marrow and human breast cancer cells, as well as leukemia cells. Additional clinical trials have shown that amifostine can protect normal tissues from the toxic effects of alkylating agents, organoplatinums, anthracyclines, taxanes, and radiation. Other laboratory and clinical investigations indicate a potential role for this cytoprotective agent in the treatment of the ineffective hematopoiesis characteristic of the myelodysplastic syndromes.


Subject(s)
Amifostine/therapeutic use , Radiation Injuries/prevention & control , Radiation-Protective Agents/therapeutic use , Amifostine/pharmacokinetics , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Clinical Trials, Phase III as Topic , Drug Therapy, Combination , Female , Humans , Leukemia/drug therapy , Leukemia/pathology , Leukemia/radiotherapy , Multicenter Studies as Topic , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/radiotherapy , Radiation-Protective Agents/pharmacokinetics , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/radiation effects
8.
Leukemia ; 12(10): 1596-602, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9766505

ABSTRACT

Amifostine (WR-2721, Ethyol) is a phosphorylated aminothiol that affords broad cytoprotection from the myelosuppressive effects of antineoplastics. To further characterize its hematopoietic activities, we investigated the effects of amifostine and its dephosphorylated metabolite, WR1065, on the in vitro growth of human bone marrow progenitors. Preincubation exposure to amifostine or WR1065 stimulated the growth of colony-forming units granulocyte, erythroid, macrophage, megakaryocyte (CFU-GEMM) and erythroid bursts (BFU-E) from bone marrow mononuclear cells in a dose-dependent fashion. Over the concentration range tested (0.1-1000 microM), pretreatment with the aminothiols enhanced formation of CFU-GEMM up to five-fold and BFU-E nine-fold, compared to a three-fold increase in myeloid colony recovery. In CD34+ selected cells, preincubation with amifostine increased formation of CFU-GEMM up to 38-fold and produced macroscopic colonies, exceeding colony number in cultures initiated with optimal concentrations of interleukin-1 (IL-1), IL-3, or kit ligand (KL). When compared with recombinant human cytokines, amifostine enhanced IL-1 and IL-3 induced colony formation, although its stimulatory effect was less than additive. In contrast, pretreatment with amifostine antagonized the stimulatory effects of KL, whereas synergy was observed with concurrent exposure. Ex vivo expansion studies showed that amifostine alone supported and augmented the production of myeloid progenitors in secondary cultures. Similarly, under cytokine-deficient conditions, amifostine promoted cell survival and delayed apoptosis as measured by nucleosome generation. These data indicate that amifostine is a novel multipotent hematopoietic stimulant that augments the formation and survival of bone marrow progenitors.


Subject(s)
Amifostine/pharmacology , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Hematopoietic Stem Cells/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/ultrastructure , Cell Division/drug effects , Cells, Cultured , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Glutathione/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/ultrastructure , Humans , Interleukin-1/pharmacology , Interleukin-3/pharmacology , Nucleosomes/drug effects , Nucleosomes/ultrastructure , Recombinant Proteins/pharmacology , Stem Cell Factor/pharmacology
9.
Eur J Cancer ; 33(10): 1693-8, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9389935

ABSTRACT

The objectives of this study were to evaluate the protective effects of amifostine against paclitaxel-induced toxicity to normal and malignant human tissues. Haematopoietic progenitor colony assays were used to establish the number of CFU-GEMM and BFU-E colonies after incubation with WR-1065 alone, Amifostine alone, paclitaxel (2.5 or 5 microM) +/- WR-1065 or amifostine. MTT and alkaline elution assays evaluated the in vitro growth inhibitory and DNA damaging effects, respectively, of paclitaxel with or without amifostine against normal human fibroblasts and human non-small cell lung cancer (NSCLC) cells. This combination was also evaluated in vivo using severe combined immune deficient (scid) mouse models of early (non-palpable tumours) and advanced (palpable tumours) human ovarian cancer. Human 2780 ovarian cancer cells were inoculated subcutaneously while paclitaxel and amifostine were administered intraperitoneally. A brief exposure (15 min) to amifostine not only protected human haematopoietic progenitor colonies from paclitaxel toxicity, but stimulated the growth of CFU-GEMM and BFU-E beyond control values. Amifostine protected normal human lung fibroblasts from paclitaxel-induced cytotoxicity and DNA single-strand breaks. However, paclitaxel cytotoxicity and DNA single-strand breaks were actually enhanced by pretreatment with amifostine in the NSCLC model. Importantly, amifostine did not interfere with paclitaxel antitumour activity even with prolonged exposure (24.5 h) of the lung cancer cells to high concentrations (1.2 mM) in vitro or following five repetitive high doses (200 mg/kg) given to scid mice with human ovarian cancer xenografts. Indeed, under certain circumstances, amifostine resulted in sensitisation of tumour cells to paclitaxel. Our results confirm previous reports of the ability of amifostine to protect normal tissues from the toxic effects of chemotherapy drugs and now extend these observations to paclitaxel.


Subject(s)
Amifostine/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cytoprotection , Ovarian Neoplasms/drug therapy , Paclitaxel/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Tumor Cells, Cultured/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Culture Techniques , Cell Death/drug effects , Female , Fibroblasts/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Lung Neoplasms/pathology , Mice , Mice, SCID , Paclitaxel/pharmacology
10.
Semin Oncol ; 23(4 Suppl 8): 35-9, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8783664

ABSTRACT

The effects of amifostine on paclitaxel-induced tumor growth delay using in vivo human ovarian cancer models were evaluated. In some mouse strains amifostine causes hypothermia and/or vasodilation, leading to increased spleen weight and ascites that can result in experimental artifacts. We found, however, that amifostine alone at 100 or 200 mg/kg intraperitoneally did not substantially alter body weight, spleen weight, or body temperature in severe combined immune-deficient (scid) mice bearing human 2780 ovarian cancer cells. In a model of minimal tumor burden (tumor cells injected subcutaneously day 0, drug treatment started day 1) scid mice receiving paclitaxel (27 mg/kg intraperitoneally) with or without amifostine had increased survival at day 76 (83% to 100%) compared with mice that did not receive paclitaxel (17% to 33%). For a model of advanced ovarian cancer, mice received tumor cell injections on day 0 and did not begin drug treatment until tumors were palpable (0.2 x 0.2 cm). Paclitaxel given for five repetitive doses significantly decreased tumor growth (P = .0001) in the advanced ovarian cancer model, and these results were the same whether or not mice received amifostine prior to each paclitaxel dose. We conclude that the scid mouse is a good model for evaluating amifostine in vivo, and that there was no evidence of amifostine-induced tumor protection in these scid mouse human ovarian cancer models. In future studies we will evaluate whether the cytoprotective effects of amifostine will allow dose escalation of paclitaxel and result in enhanced antitumor effects.


Subject(s)
Amifostine/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Disease Models, Animal , Mice, SCID , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Drug Therapy, Combination , Female , Humans , Mice , Paclitaxel/antagonists & inhibitors , Radiation-Protective Agents/therapeutic use , Tumor Cells, Cultured
11.
Semin Oncol ; 23(4 Suppl 8): 58-63, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8783669

ABSTRACT

Controlled clinical trials indicate that amifostine (WR-2721, Ethyol) confers protection from the cumulative hematologic toxicities associated with alkylating agents and organoplatinums. To determine whether amifostine protects primitive hematopoietic progenitors from the cytotoxicity of functionally diverse antineoplastics, formation of the multipotent hematopoietic progenitors colony-forming units-granulocyte-erythroid, macrophage, megakaryocyte (CFU-GEMM) and erythroid burst-forming units (BFU-E) was evaluated using a clonogenic progenitor inhibition assay. Bone marrow mononuclear cells from normal donors were subjected to a 15-minute exposure of medium, amifostine (500 micromol/L), or WR-1065 (100 micromol/L at concentrations approximating peak plasma levels, washed twice, then treated with the antineoplastic for 1 to 6 hours. Colony growth was scored after 14 days of incubation. Amifostine conferred protection against a broader range of antineoplastics than did WR-1065. Amifostine protected CFU-GEMM against cytotoxicity from daunorubicin, mitoxantrone, and paclitaxel (range, 1.29- to 9.57-fold) (P < .05) but did not afford protection against cisplatin, diaziquone, or thiotepa. Similarly, amifostine protected BFU-E against toxicity from doxorubicin, mitoxantrone, paclitaxel, cisplatin, and diaziquone, yielding 3.4- to 65-fold greater colony recovery compared with controls. The high degree of cytoprotection afforded by amifostine derived largely from stimulation of progenitor growth at sublethal chemotherapy concentrations. In the absence of antineoplastic exposure, preincubation with amifostine or WR-1065 enhanced the colony-forming capacity of bone marrow progenitors from six normal donors, increasing recovery of CFU-GEMM and BFU-E up to sevenfold. We conclude that amifostine protects primitive hematopoietic progenitors from a wide range of antineoplastics. This broad hemoprotective effect derives in part from inherent trophic effects on progenitor growth and survival.


Subject(s)
Amifostine/pharmacology , Antineoplastic Agents/adverse effects , Hematopoietic Stem Cells/drug effects , Mercaptoethylamines/pharmacology , Bone Marrow Cells , Cells, Cultured , Humans
12.
Clin Cancer Res ; 2(7): 1107-14, 1996 Jul.
Article in English | MEDLINE | ID: mdl-9816275

ABSTRACT

We conducted a combined biochemical modulation trial of N-(phosphonacetyl)-L-aspartate (PALA), dipyridamole (DP), and fluorouracil (5-FU) in patients with cancer. Eighty-eight patients with advanced cancer were entered into this Phase I trial. During the first part of the study, four doses of PALA (125, 250, 500, and 1000 mg/m2, administered on day 1) were evaluated to determine the PALA dose with maximal suppression of aspartate transcarbamylase (ATCase) activity that was clinically tolerable. Patients were randomized to receive DP (or no DP), 50 mg/m2, p.o. every 6 h on days 1-6, and all patients received 5-FU, 400 mg/m2, by bolus administration on days 2-5. Prior to and during therapy, WBCs were collected and assayed for ATCase activity. After the maximally tolerated PALA dose with 400 mg/m2 5-FU +/- 50 mg/m2 DP was defined, the 5-FU dose was escalated using the same administration schedule of 5-FU, PALA, and DP. The dose of 5-FU was escalated by 25% in each of the DP cohorts until dose-limiting toxicity was reached. ATCase activity was inhibited in a dose-dependent manner with PALA doses of 125, 250, 500, and 1000 mg/m2, resulting in 0, 13, 17, and 49% inhibition of ATCase activity. Only at the higher PALA doses (i.e., 500 and 1000 mg/m2) was ATCase activity suppressed during days 2-5, but the activity returned to pretreatment levels by day 15. Based on the clinical tolerance and significant suppression of ATCase activity, a PALA dose of 500 mg/m2 was selected for the 5-FU dose escalation phase. At a 5-FU dose of 625 mg/m2, dose-limiting toxicity (leukopenia, stomatitis, and diarrhea) occurred in both DP cohorts. We recommend that for this monthly treatment schedule, 500 mg/m2 PALA and 500 mg/m2 5-FU, with or without 50 mg/m2 DP, be used in subsequent Phase II trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Aspartate Carbamoyltransferase/antagonists & inhibitors , Aspartic Acid/administration & dosage , Aspartic Acid/adverse effects , Aspartic Acid/analogs & derivatives , Dipyridamole/administration & dosage , Dipyridamole/adverse effects , Dose-Response Relationship, Drug , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Leukocytes/enzymology , Male , Middle Aged , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/adverse effects , Phosphonoacetic Acid/analogs & derivatives , Uridine Triphosphate/metabolism
13.
Int J Radiat Oncol Biol Phys ; 34(5): 1073-9, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8600090

ABSTRACT

PURPOSE: N-(phosphonacetyl)-L-aspartic acid (PALA) is a transition- state inhibitor of L-aspartate transcarbamylase, which catalyses the biosynthesis of carbamyl-L-aspartate in the de novo pyrimidine biosynthetic pathway. 5-Bromodeoxyuridine (BrdUrd) is known to be a potent radiosensitizer of proliferating cells when it is incorporated into DNA. The experiments described herein were performed to test the hypothesis that depletion of cellular pyrimidine precursors by PALA may increase both the incorporation of BrdUrd into DNA and the sensitivity of these cells to the cytotoxic effect of radiation. METHODS AND MATERIALS: The effect of PALA concentration and exposure time on the incorporation of BrdUrd into the DNA of exponentially growing BG-1 human ovarian carcinoma cells was determined. BG-1 cells exposed to the most effective PALA + BrdUrd treatment schedule were then irradiated to determine if PALA could enhance the radiosensitization already achieved by pretreatment with BrdUrd alone. RESULTS: A 72-h exposure to PALA (> or = 25 microM) delayed the growth of human ovarian adenocarcinoma BG-1 cells by 40% compared to that of the untreated control cells. Using a clonogenic assay, the IC50 for a 72-h PALA exposure was approximately 25 microM and the cell killing efficiency was dependent on both the concentration and duration of the exposure. A 72-h exposure to 25 microM PALA produced approximately a 90% decrease in the intracellular uridine-5'-triphosphate (UTP) and cytidine-5'-triphosphate (CTP) levels, but had no effect on the intracellular adenosine-5'-triphosphate (ATP) level. This decrease in the UTP and CTP pools promoted a fivefold increase in the incorporation of [3H]BrdUrd into the DNA of BG-1 cells. The most effective treatment schedule involved a 72-h time course, consisting of a 48-h pretreatment with PALA alone, followed by an additional 24-h treatment with both PALA and BrdUrd. The two agent treatments, PALA (25 microM) + BrdUrd (16 microM), PALA (25 microM) + radiation (6 Gy), and BrdUrd (16 microM) + radiation (6 Gy) produced a 2.1-, 7.4-, and 13.2-fold increase in cytotoxicity, respectively, over that expected if the interaction between the two agents was independent and additive. The most effective three-agent treatment schedule consisting of PALA, BrdUrd, and radiation resulted in a greater than 30-fold increase in cytotoxicity over that expected if the interactions and the three agents were additive (p < 0.05). CONCLUSIONS: These data indicate that PALA alone enhances radiation cytotoxicity and further enhances the radiosensitization already achieved with the halogenated pyrimidines. These effects could be clinically beneficial.


Subject(s)
Adenocarcinoma/radiotherapy , Antimetabolites, Antineoplastic/pharmacology , Aspartic Acid/analogs & derivatives , Bromodeoxyuridine/metabolism , DNA, Neoplasm/metabolism , Ovarian Neoplasms/radiotherapy , Phosphonoacetic Acid/analogs & derivatives , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aspartic Acid/pharmacology , Cell Division/drug effects , Female , Humans , Nucleotides/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphonoacetic Acid/pharmacology , Time Factors , Tumor Stem Cell Assay
14.
Eur J Cancer ; 32A Suppl 4: S5-16, 1996.
Article in English | MEDLINE | ID: mdl-8976816

ABSTRACT

Administered prior to cytotoxic chemotherapy or radiation, the aminothiol amifostine provides broad-spectrum cytoprotection of various normal tissues without attenuating antitumour response. The basis for the selectivity of action resides in the anabolism of amifostine at the normal tissue site by membrane-bound alkaline phosphatase. Dephosphorylation to the free thiol, WR-1065, is followed by rapid uptake into normal tissues by a carrier mediated, facilitated diffusion process; in contrast, uptake into tumour tissue is slow to negligible. Preclinical studies have shown that pretreatment with amifostine provides protection of normal tissues from the cytotoxic effects of alkylating agents, organoplatinums, anthracyclines, taxanes and radiation. Normal tissues protected include bone marrow, kidney, neural tissues, the heart, intestinal crypt cells and pulmonary tissues. Additionally, the mutagenic and carcinogenic effects of these modalities are also attenuated. With respect to bone marrow, preclinical studies have shown significant protection of progenitor cells that give rise to the red and white cells and platelets. Comparative in vitro and in vivo studies using murine and human tumour xenografts show no decrease of antitumour effects of these same therapies despite the protection of normal organs. The unique preclinical profile of amifostine serves as a model for the clinical development programme for this important new broad-spectrum cytoprotective agent.


Subject(s)
Amifostine/therapeutic use , Antineoplastic Agents/adverse effects , Neoplasms, Experimental/drug therapy , Amifostine/metabolism , Animals , Antineoplastic Agents/antagonists & inhibitors , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/radiotherapy , Radiation Injuries/prevention & control , Radiation-Protective Agents/therapeutic use , Radiotherapy/adverse effects , Rats
15.
Eur J Cancer ; 32A Suppl 4: S17-20, 1996.
Article in English | MEDLINE | ID: mdl-8976817

ABSTRACT

Amifostine (WR-2721, Ethyol), a phosphorylated thiol, demonstrates the unique ability to protect normal but not tumour tissue from cytotoxic damage induced by radiation therapy and chemotherapy. This study tested the effect of amifostine's active metabolite, the free thiol, WR-1065, on the cytotoxicity of standard anticancer drugs against human A2780 ovarian and MCF7 breast cancer cell lines in vitro, using the well-characterised sulphorhodamine B assay. 50% inhibitory concentration (IC50) values were determined for each of 16 different anticancer drugs in the presence and absence of the highest nontoxic dose of WR-1065 from concentration-response curves constructed in triplicate and based on 18 replicate cell culture plates for each tested drug concentration. Pretreatment with WR-1065 had no statistically significant effect on the IC50 value of any of the 16 drugs tested against either the A2780 or MCF7 human tumour cells. These data expand upon previous reports showing that amifostine does not protect tumours from the cytotoxic effects of anticancer agents. The ability of amifostine to protect against dose-limiting toxicity to a variety of normal tissues without protection of tumour should enhance the efficacy ratio of a wide range of standard anticancer drugs.


Subject(s)
Antineoplastic Agents/antagonists & inhibitors , Breast Neoplasms/pathology , Mercaptoethylamines/pharmacology , Ovarian Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Tumor Cells, Cultured/drug effects
16.
Invest New Drugs ; 14(3): 249-56, 1996.
Article in English | MEDLINE | ID: mdl-8958179

ABSTRACT

In the bench to bedside development of drugs to treat patients with cancer, the common guide to dose and schedule selection is toxicity to normal organs patterned after the preclinical profile of the drug. An understanding of the cellular pharmacology of the drug and specifically the cellular targets linked to the drug's effect is of substantial value in assisting the clinical investigator in selecting the proper dose and schedule of drug administration. The clinical development of ara-C for the treatment of acute myeloid leukemia (AML) provides a useful paradigm for the study of this process. An understanding of the cellular pharmacology, cytokinetics and pharmacokinetics of ara-C in leukemic mice showed substantial schedule-dependency. Exposure to high doses for a short duration (C x t) resulted in a palliative therapeutic outcome. In marked contrast, exposure to lower doses for a protracted period (c x T) was curative. Clinical use of ara-C in patients with AML patterned after the murine experience, c x T approach, has been of limited benefit in terms of long-term disease-free survival. Studies with human leukemia blasts from patients have shown that for the majority of patients, the initial rate-limiting step is membrane transport, the characteristics of which are substantially affected by extracellular drug concentration (dose). This pharmacologic impediment is eliminated with the blood levels attained during the infusion of gram doses (1-3 gm/m2) of the drug (high-dose ara-C, HiDaC) for shorter periods of time, a C x t approach. Clinical confirmation of these pharmacologic observations is evident in the therapeutic efficacy of HiDaC in patients with relapsed or SDaC-refractory acute leukemia. This is further emphasized by the significantly improved leukemia-free survival of patients with AML treated with HiDaC intensification during remission compared to those patients treated with milligram doses typical of SDaC protocols. Thus, the identification and monitoring of important parameters of drug action in tumors during the course of a clinical trial can be of substantial assistance in optimizing drug dose and schedule so as to attain the best therapeutic index.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Cytarabine/therapeutic use , Leukemia L1210/drug therapy , Leukemia, Myeloid/drug therapy , Acute Disease , Animals , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Randomized Controlled Trials as Topic
17.
Blood ; 86(7): 2849-55, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7670119

ABSTRACT

One of the principal challenges of cancer chemotherapy is the relative inability of most anticancer drugs to distinguish between normal and neoplastic tissues. Consequently, a broad range of toxicities are experienced by patients, especially myelosuppression. Amifostine, a phosphorylated aminothiol, increases the selectivity of specific anticancer drugs for neoplastic cells by protecting normal tissues. One potential application of this protector is during bone marrow purging to selectively remove contaminating cancer cells. This study took normal or leukemic marrow from human subjects and evaluated the ability of amifostine to selectively protect normal bone marrow progenitor cells versus leukemic progenitor cells from the cytotoxic effect of mafosfamide. The dose response of mafosfamide amifostine on leukemia colony-forming units or normal marrow progenitor cells was determined and the LD95 was calculated. Amifostine pretreatment resulted in a statistically significant protection of granulocyte-macrophage colony-forming units and erythroid blast-forming units from the toxicity of mafosfamide (P = .031). Thus, amifostine protection of normal marrow progenitor cells allows a higher LD95 concentration of mafosfamide to be used in ex vivo purging. In contrast, amifostine pretreatment increased the cytotoxicity of mafosfamide on the fresh human leukemia progenitor cells (P = .006). The dual effect of amifostine protection of normal marrow progenitor cells coupled with amifostine-induced sensitization of the leukemia cells increases the possible cell-kill of leukemic stem cells. With amifostine pretreatment, at the LD95 concentrations of mafosfamide for marrow progenitor cells, there was an estimated 6 log increase in cell-kill of the leukemia cells. This selective cell-kill offers the potential for lowering the incidence of leukemic relapse, while preserving more normal stem cells for autologous transplantation.


Subject(s)
Amifostine/pharmacology , Antineoplastic Agents/therapeutic use , Bone Marrow Purging/methods , Cyclophosphamide/analogs & derivatives , Leukemia/pathology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Bone Marrow/pathology , Bone Marrow Transplantation , Cell Death , Cyclophosphamide/adverse effects , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Hematopoietic Stem Cells/drug effects , Humans , Leukemia/therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transplantation, Autologous
18.
Cancer Chemother Pharmacol ; 36(5): 425-30, 1995.
Article in English | MEDLINE | ID: mdl-7634384

ABSTRACT

The pharmacokinetics of cytarabine (ara-C) were determined in 265 patients with acute myeloid leukemia (AML) receiving ara-C (200 mg/m2 per day for 7 days as a continuous infusion) and daunorubicin during induction therapy. The mean (standard deviation) ara-C concentration at steady-state (Css) and systemic clearance (Cl) were 0.30 (0.13) microM and 134 (71) l/h per m2 respectively. Males had a significantly faster ara-C Cl (139 vs 131 l/h per m2, P = 0.025) than females. Significant correlations were noted between ara-C Cl and the pretreatment, peripheral white blood cell count (P = 0.005) and pretreatment blast count (P = 0.020). No significant differences in ara-C Css or Cl were noted in patients achieving complete remission compared with those failing therapy (P = 0.315, P = 0.344, respectively). No significant correlations were observed between ara-C pharmacokinetic parameters and several indices of patient toxicity. Our findings indicate that variability in ara-C disposition in plasma at this dosage level does not correlate with remission status or toxicity in patients with AML receiving initial induction therapy with ara-C and daunorubicin.


Subject(s)
Cytarabine/pharmacokinetics , Leukemia, Myeloid/blood , Acute Disease , Adolescent , Adult , Aged , Cytarabine/therapeutic use , Female , Humans , Leukemia, Myeloid/drug therapy , Male , Middle Aged , Treatment Outcome
19.
Eur J Cancer ; 31A Suppl 1: S8-13, 1995.
Article in English | MEDLINE | ID: mdl-7577096

ABSTRACT

The clinical trials described in this review indicate that amifostine protects normal tissues from the toxicities of various antitumour regimens. In a controlled trial, pretreatment with amifostine reduced the frequency of cyclophosphamide-induced neutropenia. Comparisons of the effects of cisplatin with and without pretreatment with amifostine indicated that patients pretreated with amifostine had fewer nephrotoxic and neurotoxic effects and tolerated higher doses of cisplatin before the onset of neurotoxic effects. In a randomised trial, patients who received amifostine prior to treatment with cyclophosphamide and cisplatin discontinued chemotherapy because of haemato, nephro- or ototoxicity less frequently than patients treated with cyclophosphamide and cisplatin alone. Tumour response rates and survival were comparable in both groups indicating that amifostine selectively protects only normal tissues. A regimen of amifostine plus cisplatin and vinblastine followed by amifostine plus radiation in patients with non-small cell lung cancer revealed a 73% response to treatment. Other studies showed that amifostine protected against late radiation toxicity to pelvic organs without interfering with the antitumour effect of radiotherapy, and decreased the haematological and mucosal toxicity of combined treatment with cisplatin and radiation therapy.


Subject(s)
Amifostine/therapeutic use , Antineoplastic Agents/adverse effects , Radiation Injuries/prevention & control , Radiation-Protective Agents/therapeutic use , Cisplatin/adverse effects , Clinical Trials as Topic , Cyclophosphamide/adverse effects , Humans , Neoplasms/drug therapy , Neoplasms/radiotherapy
20.
Semin Oncol ; 21(5 Suppl 11): 8-15, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7973776

ABSTRACT

The rationale for the clinical trials with amifostine (Ethyol, US Bioscience, Inc, West Conshohocken, PA) is based on an extensive body of preclinical data coming from many laboratories around the world. Initial clinical trials centered on the hematotoxicity produced by cyclophosphamide, carboplatin, and cisplatin; hematotoxicities and mucosal toxicities from radiation therapy; and nephrotoxicity, neurotoxicity, and ototoxicity from cisplatin.


Subject(s)
Amifostine/therapeutic use , Neoplasms/therapy , Amifostine/adverse effects , Antineoplastic Agents/adverse effects , Cell Survival/drug effects , Cell Survival/radiation effects , Clinical Trials as Topic , Female , Humans , Male , Neoplasms/drug therapy , Neoplasms/radiotherapy , Neutropenia/chemically induced , Neutropenia/drug therapy , Ovarian Neoplasms/drug therapy , Radiation Injuries/prevention & control , Rectal Neoplasms/radiotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...