1,218 results on '"Porfiromycin"'
Search Results
2. Porfiromycin Used as an Adjuvant to Radiation Therapy in Postoperative Head and Neck Cancer Patients
- Published
- 2014
3. Pharmacokinetic Study of Porfiromycin in Head and Neck Cancer and Other Cancer Patients With Solid Tumors
- Published
- 2014
4. Radiation Therapy Plus Porfiromycin in Treating Patients With Stage III or Stage IV Head and Neck Cancer
- Published
- 2010
5. Mitomycins and Porfiromycin: Chemical Mechanism of Activation and Cross-linking of DNA
- Author
-
Iyer, V. N. and Szybalski, Waclaw
- Published
- 1964
6. Preclinical Studies of Porfiromycin as an Adjunct to Radiotherapy
- Author
-
Rockwell, Sara, Keyes, Susan R., and Sartorelli, Alan C.
- Published
- 1988
- Full Text
- View/download PDF
7. Cyclic disulfide C8 iminoporfiromycin: nucleophilic activation of a porfiromycin.
- Author
-
Lee SH and Kohn H
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Hypoxia, Cell Line, Tumor, DNA Adducts chemistry, Imines pharmacology, Porfiromycin analogs & derivatives, Porfiromycin pharmacology, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Imines chemical synthesis, Imines chemistry, Porfiromycin chemical synthesis, Porfiromycin chemistry
- Abstract
The clinical success of mitomycin C (1) and its associated toxicities and resistance have led to efforts to prepare semisynthetic analogues (i.e., KW-2149 (3), BMS-181174 (4)) that have improved pharmacological profiles. In this study, we report the preparation and evaluation of the novel 7-N-(1'-amino-4',5'-dithian-2'-yl)porfiromycin C(8) cyclized imine (6) and its reference compound, 7-N-(1'-aminocyclohex-2'-yl)porfiromycin C(8) cyclized imine (13). Porfiromycin 6 contains a disulfide unit that, upon cleavage, may provide thiol(s) that affect drug reactivity. We demonstrated that phosphines dramatically accelerated 6 activation and solvolysis in methanolic solutions ("pH 7.4") compared with 13. Porfiromycins 6 and 13 efficiently cross-linked EcoRI-linearized pBR322 DNA upon addition of Et3P. We found enhanced levels of interstrand cross-link (ISC) adducts for 6 and 13 compared with porfiromycin (7) and that 6 was more efficient than 13. The large Et3P-mediated rate enhancements for the solvolysis of 6 compared with 13 and a N(7)-substituted analogue of 1, and the increased levels of ISC adducts for 6 compared with 13 and 7 are attributed to a nucleophile-assisted disulfide cleavage process that permits porfiromycin activation and nucleophile (MeOH, DNA) adduction. The in vitro antiproliferative activities of 6 and 13 using the A549 tumor cell line (lung adenocarcinoma) were determined under aerobic and hypoxic conditions and then compared with 7. Both 6 and 13 were more cytotoxic than 7, with 13 being more potent than 6. The C(8) iminoporfiromycins 6 and 13 displayed anticancer profiles similar to 3.
- Published
- 2004
- Full Text
- View/download PDF
8. Isolation and identification of urinary metabolites of porfiromycin in dogs and humans.
- Author
-
Lang W, Mao J, Doyle TW, and Almassian B
- Subjects
- Animals, Antibiotics, Antineoplastic chemistry, Antibiotics, Antineoplastic metabolism, Antibiotics, Antineoplastic urine, Chromatography, High Pressure Liquid, Dogs, Humans, Magnetic Resonance Spectroscopy, Male, Mass Spectrometry, Porfiromycin chemistry, Porfiromycin isolation & purification, Radiopharmaceuticals chemistry, Radiopharmaceuticals isolation & purification, Tritium, Porfiromycin urine, Radiopharmaceuticals urine
- Abstract
Porfiromycin (PM), a bioreductive alkylating agent, is currently under development for the treatment of head and neck cancers as an adjunct to radiation therapy in phase III clinical trials. After i.v. administration of a single dose of PM to patients at 40 mg/m2, urinary metabolites were isolated by HPLC and identified by atmospheric pressure chemical ionization mass spectrometry. In dogs, [methyl-3H]PM was administered i.v. to three Beagle dogs at a single dose of 2 mg/kg. Urinary excretion of radioactivity and PM at different times was determined by liquid scintillation counting and by HPLC, respectively. An average of 48.0% of total radioactivity given to the dogs was cumulatively excreted in urine over a period of 7 days. Unchanged parent drug excreted in urine accounted for 10.8% of the administered dose over the same period of time. The results indicated that the majority of excreted dose in dog urine was in the form of metabolites. Three phase I and four phase II metabolites of PM were identified in human and dog urine. The phase I metabolites are 2-methylamino-7-aminomitosene, 1,2-cis and 1,2-trans-1-hydroxy-2-methylamino-7-aminomitosenes. The phase II metabolites are a pair of isomeric N-acetylcysteine S-conjugates and a pair of isomeric cysteine S-conjugates of mitosenes at the C-1 and C-10 positions. Most of the identified metabolites were confirmed by comparison with synthetic reference standards using HPLC and liquid chromatography/mass spectrometry (LC/MS). The identification of mercapturic acids and cysteine S-conjugates in urine indicates that the metabolism of PM may be through GSH conjugation.
- Published
- 2000
9. Isolation and identification of metabolites of porfiromycin formed in the presence of a rat liver preparation.
- Author
-
Lang W, Mao J, Wang Q, Niu C, Doyle TW, and Almassian B
- Subjects
- Acetylcysteine metabolism, Animals, Antibiotics, Antineoplastic isolation & purification, Biotransformation, Chromatography, High Pressure Liquid, Cysteine metabolism, Liver chemistry, Mass Spectrometry, Nuclear Magnetic Resonance, Biomolecular, Porfiromycin pharmacokinetics, Rats, Rats, Sprague-Dawley, Spectrophotometry, Ultraviolet, Antibiotics, Antineoplastic metabolism, Liver metabolism, Porfiromycin isolation & purification, Porfiromycin metabolism
- Abstract
The isolation and identification of the major metabolites of porfiromycin formed in the presence of a rat liver preparation under aerobic conditions were performed with high-performance liquid chromatography and electrospray ionization mass spectrometry. Porfiromycin was extensively metabolized by the rat liver preparation in an aqueous 0.1 M potassium phosphate buffer (pH 7.4) containing an NADPH generating system at 37 degrees C. A total of eight metabolites was identified as mitosene analogs. Of these, three primary metabolites are 2-methylamino-7-aminomitosene, 1,2-cis and 1,2-trans-1-hydroxy-2-methylamino-7-aminomitosene, which are consistent with those previously observed in hypoxia using purified rat liver NADPH-cytochrome c reductase. Interestingly, 2-methylamino-7-aminomitosene is a reactive metabolite, which undergoes further activation at the C-10 position by the loss of carbamic acid and then links with the 7-amino group of the primary metabolites to yield two dimeric adducts. In addition, three phosphate adducts, 10-decarbamoyl-2-methylamino-7-aminomitosene-10-phosphate, 1,2-cis and 1,2-trans-2-methylamino-7-aminomitosene-1-phosphate, were also identified in the incubation system. The configurations of the diastereoisomeric metabolites were determined with (1)HNMR and phosphatase digestion. On the basis of the metabolite profile, we propose in vitro metabolic pathways for porfiromycin. The findings provide direct evidence for understanding the reactive nature and hepatic metabolism of the drug currently in phase III clinical trials., (Copyright 2000 Wiley-Liss, Inc.)
- Published
- 2000
- Full Text
- View/download PDF
10. Bioreductive alkylating agent porfiromycin in combination with radiation therapy for the management of squamous cell carcinoma of the head and neck.
- Author
-
Haffty BG, Son YH, Wilson LD, Papac R, Fischer D, Rockwell S, Sartorelli AC, Ross D, Sasaki CT, and Fischer JJ
- Subjects
- Antibiotics, Antineoplastic adverse effects, Carcinoma, Squamous Cell mortality, Combined Modality Therapy, Dose Fractionation, Radiation, Follow-Up Studies, Head and Neck Neoplasms mortality, Humans, Mitomycin adverse effects, Mitomycin therapeutic use, Porfiromycin adverse effects, Survival Rate, Time Factors, Antibiotics, Antineoplastic therapeutic use, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell radiotherapy, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms radiotherapy, Porfiromycin therapeutic use
- Abstract
Porfiromycin (methyl mitomycin C) has been shown in laboratory studies to have increased preferential cytotoxicity to hypoxic cells and therefore may provide enhanced therapeutic efficacy over mitomycin C when used in combination with radiation therapy (RT). The purpose of the two clinical studies reported here is to evaluate the concomitant use of porfiromycin with RT in the management of squamous cell carcinoma of the head and neck. Between October 1989 and July 1992, 21 patients presenting with locally advanced stage III/IV squamous cell carcinoma of the head and neck were entered into a phase I toxicity trial evaluating porfiromycin as an adjunct to RT. Patients were eligible if they had biopsy documented squamous cell carcinoma of the head and neck with a low probability of cure by conventional means. Patients were treated with standard fractionated daily RT to a total median dose of 63 Gy, with porfiromycin administered on days 5 and 47 of the course of RT. Upon completion of this phase I trial, a phase III trial was initiated in November 1992 randomizing patients with squamous cell carcinoma of the head and neck to RT with mitomycin C vs. RT with porfiromycin. There is no radiation only arm in this current trial. To date, 75 patients have been entered on this trial and acute toxicity data are available on 67 patients (34 porfiromycin, 31 mitomycin C) who have completed their entire course of treatment. Median follow-up of the 21 patients enrolled in the phase I porfiromycin trial is 58.5 months. Of the 21 patients, 5 were treated at a dose of 50 mg/M2, 4 at 45 mg/M2, and the final 12 at 40 mg/M2, which appeared to result in acceptable acute hematological and nonhematological toxicities. As of December 1995, 14 of the 21 patients have died with disease and 7 remain alive and free of disease, resulting in a 5-year actuarial survival of 32%. Of the patients enrolled to date in the phase III randomized trial of mitomycin C vs. porfiromycin, there have been no statistically significant differences between the two arms with respect to white blood cell count (WBC), platelet, or hemoglobin nadirs. Acute nonhematological toxicities including mucositis, epidermitis, odynophagia, and nausea have also been comparable. Two patients in this current randomized trial died during treatment, apparently of nondrug-related causes. We conclude that the bioreductive alkylating agent porfiromycin has demonstrated an acceptable toxicity profile to date. Final analysis of the phase I trial, which revealed a 5-year no evidence of disease survival rate of 32% in patients with locally advanced disease and a low probability of cure, appears encouraging. We anticipate completion of the current ongoing trial comparing mitomycin C to porfiromycin in the next 2 years. Further investigations, including large-scale multiinstitutional trials employing bioreductive alkylating agents or other hypoxic cell cytotoxins as adjuncts to RT, are warranted.
- Published
- 1997
- Full Text
- View/download PDF
11. Preferential kill of hypoxic EMT6 mammary tumor cells by the bioreductive alkylating agent porfiromycin.
- Author
-
Sartorelli AC, Belcourt MF, Hodnick WF, Keyes SR, Pritsos CA, and Rockwell S
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacokinetics, Cell Survival drug effects, Free Radicals metabolism, Hydroxyl Radical metabolism, Mammary Neoplasms, Experimental metabolism, Mice, Mice, Inbred BALB C, Mitomycin pharmacokinetics, Mitomycin pharmacology, Molecular Structure, NADPH-Ferrihemoprotein Reductase, Oxidoreductases metabolism, Oxygen Consumption, Porfiromycin pharmacokinetics, Antibiotics, Antineoplastic pharmacology, Cell Hypoxia, Mammary Neoplasms, Experimental pathology, Porfiromycin pharmacology
- Abstract
Hypoxic cells in solid tumors represent a therapeutically resistant population that limits the curability of many solid tumors by irradiation and by most chemotherapeutic agents. The oxygen deficit, however, creates an environment conducive to reductive processes; this results in a major exploitable difference between normal and neoplastic tissues. The mitomycin antibiotics can be reductively activated by a number of oxidoreductases, in a process required for the production of their therapeutic effects. Preferential activation of these drugs under hypoxia and greater toxicity to oxygen-deficient cells than to their oxygenated counterparts are obtained in most instances. The demonstration that mitomycin C and porfiromycin, used to kill the hypoxic fraction, in combination with irradiation, to eradicate the oxygenated portion of the tumor, produced enhanced cytodestructive effects on solid tumors in animals has led to the clinical evaluation of the mitomycins in combination with radiation therapy in patients with head and neck cancer. The findings from these clinical trials have demonstrated the value of directing a concerted therapeutic attack on the hypoxic fraction of solid tumors as an approach toward enhancing the curability of localized neoplasms by irradiation.
- Published
- 1995
- Full Text
- View/download PDF
12. Porfiromycin as an adjunct to radiotherapy in young and old mice.
- Author
-
Rockwell S, Hughes CS, Keyes SR, Sartorelli AC, and Kennedy KA
- Subjects
- Animals, Carcinoma metabolism, Combined Modality Therapy, Evaluation Studies as Topic, Lissamine Green Dyes, Mammary Neoplasms, Experimental metabolism, Mice, Mice, Inbred BALB C, Neoplasm Transplantation, Porfiromycin metabolism, Aging physiology, Carcinoma drug therapy, Carcinoma radiotherapy, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental radiotherapy, Porfiromycin therapeutic use
- Abstract
Radiobiological data and measurements with O2 microelectrodes show that EMT6 tumors implanted into aged mice have a higher proportion of radioresistant, hypoxic cells than do tumors implanted into young adult animals; radiation is less effective in killing cells in tumors in old mice than in tumors in young adult mice. The studies reported here examine the effects of porfiromycin (POR), a bioreductive alkylating agent shown previously to be preferentially toxic to hypoxic EMT6 cells in vitro and in solid tumors in young adult mice. POR was effective in attacking the hypoxic cells of tumors in aged mice; regimens combining POR with x-rays overcame the radioresistance of tumors in the old animals. Comparisons of the distribution of 3H-labeled POR in young and old mice showed that tumors in aged mice had a slightly larger proportion of areas with necrotic features, which bound higher levels of tritiated POR than did healthy tumor regions without necrotic features. Studies of histology, lissamine green distributions, binding of tritiated POR, and radiation and POR cytotoxicity suggested that tumors in old mice contained a larger proportion of poorly perfused tumor cells, and that cells in these regions were resistant to radiation and sensitive to POR. Studies of the distribution of POR in normal tissues and of the toxicity of POR to bone marrow progenitor cells (CFU-GM) revealed no differences between young and old animals, showing that the differences observed in tumors reflected differences in the microenvironments within the tumors, rather than differences in the processing of drug in young and old animals.
- Published
- 1993
- Full Text
- View/download PDF
13. The role of NAD(P)H:quinone oxidoreductase in mitomycin C- and porfiromycin-resistant HCT 116 human colon-cancer cells.
- Author
-
Pan SS, Akman SA, Forrest GL, Hipsher C, and Johnson R
- Subjects
- 2,6-Dichloroindophenol metabolism, Carbon Radioisotopes, Cell Extracts, Cell Size physiology, Cell Survival drug effects, Cell Survival physiology, Colonic Neoplasms drug therapy, Colonic Neoplasms genetics, Drug Resistance, Drug Screening Assays, Antitumor, Humans, Intracellular Fluid metabolism, Microsomes metabolism, Mitomycin metabolism, NAD(P)H Dehydrogenase (Quinone) genetics, Oxidation-Reduction, Porfiromycin pharmacokinetics, RNA, Messenger genetics, Tissue Distribution, Transcription, Genetic genetics, Tumor Cells, Cultured drug effects, Vitamin K metabolism, Colonic Neoplasms enzymology, Mitomycin pharmacology, NAD(P)H Dehydrogenase (Quinone) physiology, Porfiromycin pharmacology
- Abstract
A mitomycin C (MMC)- and porfiromycin (PFM)-resistant subline of the HCT 116 human colon-cancer cell line was isolated after repeated exposure of HCT 116 cells to increasing concentrations of MMC under aerobic conditions. The MMC-resistant subline (designated HCT 116-R30A) was 5 times more resistant than the parent cells to MMC and PFM under aerobic conditions. Both the MMC-resistant cells and the parent HCT 116 cells accumulated similar amounts of PFM by passive diffusion, but levels of macromolecule-bound PFM were about 50% lower in the resistant cell line, implying a decrease in PFM reductive activation in the resistant cells. The finding that microsomes from either sensitive or resistant cells showed an equal ability to reduce MMC and PFM indicated that the activity of NADPH cytochrome P-450 reductase (EC 1.6.2.4) was not changed in the resistant subline. Soluble extracts of HCT 116 cells reduced MMC and PFM more effectively at pH 6.1, and NADH and NADPH were utilized equally well as electron donors under both aerobic and anaerobic conditions. These data suggest that quinone reductase (EC 1.6.99.2; DT-diaphorase) in soluble extracts is responsible for the reduction of MMC. Quinone reductase activities in soluble extracts of HCT 116-R30A cells for the reduction of dichlorophenol indophenol (DCPIP) and menadione-cytochrome c at optimal pHs were decreased by 95% as compared with those obtained in parent cells. However, the MMC-reducing activity of HCT 116-R30A soluble extracts was only 50% lower than that of the parent cell extracts. The kinetic constants (Km, Vmax) found for quinone reductase in the two cell lines with respect to the substrates DCPIP and menadione differed. Two species of mRNA for quinone reductase (2.7 and 1.2 kb) were detected in both cell lines, and there was no detectable difference between parent and resistant cells in the steady-state level of either of these mRNA species. Furthermore, incubation with the quinone reductase inhibitor dicoumarol rendered HCT 116 cells more resistant to MMC. Alteration of the quinone reductase activity in HCT 116-R30A cells appears to be the mechanism responsible for their resistance to MMC and PFM.
- Published
- 1992
- Full Text
- View/download PDF
14. Activity of C-7 substituted cyclic acetal derivatives of mitomycin C and porfiromycin against hypoxic and oxygenated EMT6 carcinoma cells in vitro and in vivo.
- Author
-
Rockwell S, Keyes SR, Loomis R, Kelley M, Vyas DM, Wong H, Doyle TW, and Sartorelli AC
- Subjects
- Animals, Antineoplastic Agents chemistry, Cell Hypoxia, Cell Survival, Combined Modality Therapy, Drug Screening Assays, Antitumor, Mice, Mice, Inbred BALB C, Porfiromycin therapeutic use, Structure-Activity Relationship, Tumor Cells, Cultured drug effects, Antineoplastic Agents therapeutic use, Mammary Neoplasms, Experimental drug therapy, Mitomycins therapeutic use, Porfiromycin analogs & derivatives
- Abstract
A series of cyclic acetal derivatives of mitomycin C (MC) and porfiromycin (POR) were tested for their ability to kill hypoxic and oxygenated EMT6 tumor cells. Amino methyl acetal and thioacetal substitutions at C-7 of MC and POR dramatically increased the cytotoxicity of the compounds to hypoxic EMT6 tumor cells in vitro but had little effect on the aerobic toxicities. In contrast, a methyl substitution at N1a markedly decreased the aerobic cytotoxicities of the compounds but did not alter the hypoxic cytotoxicities. The POR acetal, BMY-42355, had the largest differential between hypoxic and aerobic cytotoxicities yet observed among MC analogs. Preliminary studies in mice showed that BMY-42355 had good antineoplastic activity when used alone or in combination with radiation and was less toxic than POR; the therapeutic ratio of this compound in these initial studies was higher than those of either MC or POR.
- Published
- 1991
- Full Text
- View/download PDF
15. Structural, conformational, and theoretical binding studies of antitumor antibiotic porfiromycin (N-methylmitomycin C), a covalent binder of DNA, by X-ray, NMR, and molecular mechanics.
- Author
-
Arora SK, Cox MB, and Arjunan P
- Subjects
- Antibiotics, Antineoplastic metabolism, Base Sequence, Chemical Phenomena, Chemistry, Physical, Magnetic Resonance Spectroscopy, Models, Molecular, Molecular Conformation, Molecular Sequence Data, Molecular Structure, Porfiromycin metabolism, Thermodynamics, X-Ray Diffraction, Antibiotics, Antineoplastic chemistry, DNA metabolism, Porfiromycin chemistry
- Abstract
X-ray, NMR, and molecular mechanics studies on antitumor antibiotic porfiromycin (C16H20N4O5), a covalent binder of DNA, have been carried out to study the structure, conformation, and theoretical interactions with DNA. The crystal structure was solved by direct methods and refined to an R value of 0.052. The configurations at C(9), C(9a), C(1), and C(2) are S, R, S, and S, except for the orientation of the aziridine ring and (carbamoyloxy)methyl side chain. The five-membered ring attached to the aziridine ring adopts an envelope conformation. The solution conformation is similar to that observed in the solid state except for the (carbamoyloxy)methyl side chain. Monovalent and cross-linked models of the drug bound to DNA have been energetically refined by using molecular mechanics. The results indicate that, in the case of monocovalent binding, the drug clearly prefers a d(CpG) sequence rather than a d(GpC) sequence. In the case of the cross-linked model there is no clear-cut preference of d(CpG) over d(GpC), indicating that the binding preference of the drug may be kinetic rather than thermodynamic.
- Published
- 1990
- Full Text
- View/download PDF
16. Porfiromycin disposition in oxygen-modulated P388 cells.
- Author
-
Pan SS
- Subjects
- Aerobiosis, Animals, Carbon Radioisotopes, Cell Hypoxia, DNA metabolism, Dealkylation, Mice, Porfiromycin pharmacology, Tumor Cells, Cultured, Leukemia P388 metabolism, Oxygen pharmacology, Porfiromycin metabolism
- Abstract
The cytotoxicity, metabolism, and DNA alkylation of porfiromycin (PFM) under aerobic and hypoxic conditions were evaluated in P388 murine leukemia cells. Clonogenic assays showed that the IC50 value for a 1-h exposure to PFM was 4 microM for aerobic cells and 0.5 microM for hypoxic cells. After a 1-h exposure to concentrations of 1, 5, and 10 microM [14C]-PFM, the accumulation of total radioactivity in hypoxic cells was 10 to 20 times that in aerobic cells. The disposition of radioactivity in cells that had been treated for 1 h with 5 microM PFM under aerobic or hypoxic conditions showed that (a) under either condition, internal free-PFM concentration equalled the external drug concentration; (b) DNA-, RNA-, and protein-bound radioactivity were at least 10 times greater in hypoxic cells than in aerobic cells; and (c) known metabolites and unidentified radioactive products were also generated in greater amounts in hypoxic cells than in aerobic cells. Thus, the increased amounts of radioactivity accumulated by hypoxic P388 cells after exposure to [14C]-PFM resulted from the accumulation of nonexchangeable protein and nucleic-acid adducts and metabolites rather than free PFM. Determinations of DNA adducts formed in P388 cells revealed five possible adducts: (1) N2-(2'-deoxyguanosyl)-7-methylaminomitosene, (2) a second monofunctional PFM-guanine adduct, (3) a PFM cross-linked dinucleotide, (4) possibly a nucleoprotein-related adduct, and (5) an unknown. We conclude that the enhancement of PFM-induced cytotoxicity by hypoxia appears to be primarily due to increased alkylation of macromolecules.
- Published
- 1990
- Full Text
- View/download PDF
17. The Binding of Porfiromycin to Deoxyribonucleic Acid
- Author
-
White, H. L. and White, J. R.
- Published
- 1965
18. Concurrent chemo-radiotherapy with mitomycin C compared with porfiromycin in squamous cell cancer of the head and neck: final results of a randomized clinical trial.
- Author
-
Haffty BG, Wilson LD, Son YH, Cho EI, Papac RJ, Fischer DB, Rockwell S, Sartorelli AC, Ross DA, Sasaki CT, and Fischer JJ
- Subjects
- Analysis of Variance, Combined Modality Therapy, Disease-Free Survival, Female, Humans, Leukopenia chemically induced, Male, Middle Aged, Porfiromycin therapeutic use, Prospective Studies, Radiotherapy Dosage, Statistics, Nonparametric, Thrombocytopenia chemically induced, Antibiotics, Antineoplastic therapeutic use, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell radiotherapy, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms radiotherapy, Mitomycin therapeutic use
- Abstract
Purpose: Previous randomized trials have shown a benefit with concurrent use of the hypoxic cell cytotoxin mitomycin C (MC) and radiation (RT) in the management of squamous cell cancer of the head and neck (SCCHN). We conducted a randomized trial comparing MC with porfiromycin (POR) in combination with RT in the management of SCCHN., Methods and Materials: Between 1992 and 1999, 128 patients with SCCHN were enrolled in this prospective randomized trial. Patients were stratified by management intent, and balanced with respect to stage and site of disease. They were randomized to receive MC (15 mg/M(2)) or POR (40 mg/M(2)) on Days 5 and 47 (or last day) of RT. Of 121 evaluable patients, 61 were randomized to MC and 60 to POR. Patients were treated with standard daily RT to a total median dose of 64 Gy over 47 days. Patients were well balanced with respect to management intent, stage, site, age, sex, hemoglobin levels, tumor grade, radiation dose, and days on treatment., Results: There were no significant differences between the two arms with respect to acute hematologic or nonhematologic toxicities. As of January 2003 with a median follow-up of 6.3 years, there have been 19 local relapses (4 MC vs. 15 POR), 21 regional relapses (7 MC vs. 14 POR), 24 distant metastases (11 MC vs. 13 POR), and 66 deaths (33 MC vs. 33 POR). MC was superior to POR with respect to 5-year local relapse-free survival (91.6% vs. 72.7%, p = 0.01), local-regional relapse-free survival (82% vs. 65.3%, p = 0.05), and disease-free survival (72.8% vs. 52.9%, p = 0.026). There were no significant differences between the two arms with respect to overall survival (49.2% vs. 54.4%) or distant metastasis-free rate (79.9% vs. 75.9%)., Conclusions: Despite promising preclinical data, and an acceptable toxicity profile, POR was inferior to MC as an adjunct to RT in the management of SCCHN. This randomized trial emphasizes the need for randomized studies to evaluate new agents in the management of SCCHN.
- Published
- 2005
- Full Text
- View/download PDF
19. Phenethyl Alcohol Synergism with Mitomycin C, Porfiromycin, and Streptonigrin
- Author
-
White, James R. and White, Helen L.
- Published
- 1964
20. Cyclic Disulfide C(8) Iminoporfiromycin: Nucleophilic Activation of a Porfiromycin
- Author
-
Harold Kohn and Sang Hyup Lee
- Subjects
chemistry.chemical_classification ,Stereochemistry ,Imine ,Antineoplastic Agents ,General Chemistry ,Biochemistry ,Cell Hypoxia ,Porfiromycin ,Catalysis ,Adduct ,DNA Adducts ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Polycyclic compound ,chemistry ,Nucleophile ,Cell Line, Tumor ,Thiol ,Animals ,Reactivity (chemistry) ,Imines ,Solvolysis - Abstract
The clinical success of mitomycin C (1) and its associated toxicities and resistance have led to efforts to prepare semisynthetic analogues (i.e., KW-2149 (3), BMS-181174 (4)) that have improved pharmacological profiles. In this study, we report the preparation and evaluation of the novel 7-N-(1'-amino-4',5'-dithian-2'-yl)porfiromycin C(8) cyclized imine (6) and its reference compound, 7-N-(1'-aminocyclohex-2'-yl)porfiromycin C(8) cyclized imine (13). Porfiromycin 6 contains a disulfide unit that, upon cleavage, may provide thiol(s) that affect drug reactivity. We demonstrated that phosphines dramatically accelerated 6 activation and solvolysis in methanolic solutions ("pH 7.4") compared with 13. Porfiromycins 6 and 13 efficiently cross-linked EcoRI-linearized pBR322 DNA upon addition of Et3P. We found enhanced levels of interstrand cross-link (ISC) adducts for 6 and 13 compared with porfiromycin (7) and that 6 was more efficient than 13. The large Et3P-mediated rate enhancements for the solvolysis of 6 compared with 13 and a N(7)-substituted analogue of 1, and the increased levels of ISC adducts for 6 compared with 13 and 7 are attributed to a nucleophile-assisted disulfide cleavage process that permits porfiromycin activation and nucleophile (MeOH, DNA) adduction. The in vitro antiproliferative activities of 6 and 13 using the A549 tumor cell line (lung adenocarcinoma) were determined under aerobic and hypoxic conditions and then compared with 7. Both 6 and 13 were more cytotoxic than 7, with 13 being more potent than 6. The C(8) iminoporfiromycins 6 and 13 displayed anticancer profiles similar to 3.
- Published
- 2004
- Full Text
- View/download PDF
21. Effect of pH on DNA alkylation by enzyme-activated mitomycin C and porfiromycin.
- Author
-
Yu F and Pan SS
- Subjects
- Alkylation, Animals, Biotransformation, Cattle, Chromatography, High Pressure Liquid, Chromatography, Thin Layer, Hydrogen-Ion Concentration, Liver enzymology, NADPH-Ferrihemoprotein Reductase metabolism, Phosphodiesterase I, Phosphoric Diester Hydrolases metabolism, Rats, Xanthine Oxidase metabolism, DNA metabolism, DNA Adducts, Mitomycin metabolism, Porfiromycin metabolism
- Abstract
DNA adduct formation by enzyme-activated antibiotics, mitomycin C (MMC) or porfiromycin (PFM), at pH 7.6 or pH 6.0 under anaerobic conditions was analyzed by a 32P-postlabeling method. Antibiotic activation by rat liver NADPH-cytochrome P-450 reductase (EC 1.6.2.4) and bovine milk xanthine oxidase (EC 1.2.3.2) produced similar results. Five 32P-labeled MMC adducts were separated by thin layer chromatography and high performance liquid chromatography from DNA alkylated at either pH. Four of the radioactive spots separated by thin layer chromatography were identified as two monofunctional monoadducts [1" alpha and 1" beta forms of N2-(2" beta,7"-diaminomitosen-1"-yl)-2'-deoxyguanylic acid], one bifunctional monoadduct [N2-(10"-decarbamoyl-2",7"-diaminomitosen-1" alpha-yl)-2'-deoxyguanylic acid], and one cross-linked adduct [N2-(2" beta,7"-diamino-10"-deoxyguanyl-N2-yl-mitosen- 1" alpha-yl)-2'-deoxyguanylic acid]. One minor radioactive spot was not identified. By comparing DNA alkylated at the two pH values, based on equal amounts of 32P radioactivity, similar amounts of cross-links were detected. However, the DNA showed different ratios of the alpha and beta isomers of the monofunctional monoadduct. Furthermore, the DNA alkylated at pH 6.0 showed more bifunctional monoadducts than did the DNA alkylated at pH 7.6. Analysis of alkylated DNA by enzyme-activated PFM showed a similar spectrum of DNA adduct formation. The effect of pH on the distribution of the five PFM-DNA adducts was similar to that observed for the five MMC-DNA adducts. The distribution of adducts in DNA alkylated at the same pH was similar irrespective of which enzyme activated MMC or PFM. The pH of the reaction during DNA and MMC interaction was the determining factor for the quantitative distribution of the adducts. This pH effect may be important for the cytotoxicity of MMC and PFM in tumor cells that have high levels of reductive enzymes with low optimal pH values.
- Published
- 1993
22. Concurrent chemo-radiotherapy with mitomycin C compared with porfiromycin in squamous cell cancer of the head and neck: Final results of a randomized clinical trial
- Author
-
James J. Fischer, Diana B. Fischer, Douglas A. Ross, Rose J. Papac, Lynn D. Wilson, Edward I. Cho, Sara Rockwell, Yung H. Son, Alan C. Sartorelli, Clarence T. Sasaki, and Bruce G. Haffty
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Mitomycin ,medicine.medical_treatment ,Disease-Free Survival ,Statistics, Nonparametric ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Prospective Studies ,Stage (cooking) ,Head and neck ,Analysis of Variance ,Chemotherapy ,Antibiotics, Antineoplastic ,Radiation ,Squamous cell cancer ,business.industry ,Mitomycin C ,Radiotherapy Dosage ,Leukopenia ,Middle Aged ,Combined Modality Therapy ,Thrombocytopenia ,Porfiromycin ,Surgery ,Radiation therapy ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,Female ,business - Abstract
Purpose Previous randomized trials have shown a benefit with concurrent use of the hypoxic cell cytotoxin mitomycin C (MC) and radiation (RT) in the management of squamous cell cancer of the head and neck (SCCHN). We conducted a randomized trial comparing MC with porfiromycin (POR) in combination with RT in the management of SCCHN. Methods and materials Between 1992 and 1999, 128 patients with SCCHN were enrolled in this prospective randomized trial. Patients were stratified by management intent, and balanced with respect to stage and site of disease. They were randomized to receive MC (15 mg/M 2 ) or POR (40 mg/M 2 ) on Days 5 and 47 (or last day) of RT. Of 121 evaluable patients, 61 were randomized to MC and 60 to POR. Patients were treated with standard daily RT to a total median dose of 64 Gy over 47 days. Patients were well balanced with respect to management intent, stage, site, age, sex, hemoglobin levels, tumor grade, radiation dose, and days on treatment. Results There were no significant differences between the two arms with respect to acute hematologic or nonhematologic toxicities. As of January 2003 with a median follow-up of 6.3 years, there have been 19 local relapses (4 MC vs. 15 POR), 21 regional relapses (7 MC vs. 14 POR), 24 distant metastases (11 MC vs. 13 POR), and 66 deaths (33 MC vs. 33 POR). MC was superior to POR with respect to 5-year local relapse-free survival (91.6% vs. 72.7%, p = 0.01), local-regional relapse-free survival (82% vs. 65.3%, p = 0.05), and disease-free survival (72.8% vs. 52.9%, p = 0.026). There were no significant differences between the two arms with respect to overall survival (49.2% vs. 54.4%) or distant metastasis–free rate (79.9% vs. 75.9%). Conclusions Despite promising preclinical data, and an acceptable toxicity profile, POR was inferior to MC as an adjunct to RT in the management of SCCHN. This randomized trial emphasizes the need for randomized studies to evaluate new agents in the management of SCCHN.
- Published
- 2005
- Full Text
- View/download PDF
23. Phase I studies of porfiromycin (NSC--56410) in solid tumors.
- Author
-
Grage TB, Weiss AJ, Wilson W, and Reynolds V
- Subjects
- Drug Evaluation, Humans, Leukopenia chemically induced, Necrosis, Porfiromycin administration & dosage, Porfiromycin toxicity, Thrombocytopenia chemically induced, Neoplasms drug therapy, Porfiromycin therapeutic use
- Abstract
Porfiromycin was given to a group of patients with a variety of solid tumors. Of 114 patients admitted to the study, 103 yielded evaluable data. The following dosage schedules were used to determine the toxicity of porfiromycin when given in multiple doses by intravenous injection: 0.2 mg/kg x 5 days, 0.3 mg/kg x 5 days, 0.35 mg/kg x 5 days, 0.4 mg/kg x 5 days, 0.24 mg/kg x 10 days and 0.6 mg/kg weekly. Toxic effects noted were mainly leukopenia, thrombocytopenia, and, when injected paravenously, local tissue necrosis. Biological effects were noted at all dosage levels and were more severe at the higher dosages. The data suggest that profiromycin administered intravenously at a dose of 0.35 mg/kg daily for 5 days results in moderate hermatological toxicity and clinical evaluation in a Phase II study at this dosage level is indicated.
- Published
- 1975
- Full Text
- View/download PDF
24. Development of new mitomycin C and porfiromycin analogues.
- Author
-
Iyengar BS, Lin HJ, Cheng L, Remers WA, and Bradner WT
- Subjects
- Animals, Dose-Response Relationship, Drug, Leukemia, Experimental drug therapy, Melanoma drug therapy, Mice, Mitomycins pharmacology, Neoplasms, Experimental drug therapy, Porfiromycin analogs & derivatives, Porfiromycin pharmacology, Structure-Activity Relationship, Antineoplastic Agents, Mitomycins chemical synthesis, Porfiromycin chemical synthesis
- Abstract
New mitomycin C and porfiromycin analogues were prepared by treating mitomycin A and N-methylmitomycin A with a variety of amines, including aziridines, allylamines, propargylamines, chloroalkylamines, hydroxyalkylamines, glycine derivatives, aralkylamines, and heterocyclic amines. All analogues were evaluated against P-388 murine leukemia and selected ones were examined for their leukopenic properties. Certain analogues were found to be superior to mitomycin C in potency, efficacy, and therapeutic ratio in the P-388 assay. The most active substituents at the mitosane 7 position included aziridine, 2-methylaziridine, propargylamine, furfurylamine, methyl glycinate, and 3-aminopyridine. Mitomycin A and the 7-aziridino, 7-(2-methylaziridino), and 3-aminopyridine analogues were less leukopenic than mitomycin C. Certain other analogues, including propargylamino and methyl glycinate, were highly leukopenic. The three compounds tested against B-16 melanoma in mice were significantly more effective than mitomycin C in this assay. Previously established structure--activity relationships were found inadequate to account for all of the new data.
- Published
- 1981
- Full Text
- View/download PDF
25. Modulation of the cytotoxicity of porfiromycin by dicoumarol in vitro and in vivo.
- Author
-
Rockwell S, Keyes SR, and Sartorelli AC
- Subjects
- Animals, Cell Line, Cell Survival drug effects, Mice, Mice, Inbred BALB C, Porfiromycin pharmacology, Porfiromycin therapeutic use, Tumor Cells, Cultured cytology, Tumor Cells, Cultured drug effects, Tumor Stem Cell Assay, Dicumarol therapeutic use, Mammary Neoplasms, Experimental drug therapy, Mitomycins toxicity, Porfiromycin toxicity
- Abstract
The effects of dicoumarol (DIC) on the cytotoxicity of porfiromycin (POR) were studied in vitro using EMT6 mammary tumor cells in monolayer cultures and in vivo using solid EMT6 tumors and bone marrow stem cells. In vitro, POR was more toxic to hypoxic EMT6 cells than to aerobic cells. Exposure of aerobic cultures to DIC protected against POR; in contrast, DIC sensitized hypoxic cells to POR. Treatment of mice with DIC produced a slight increase in the toxicity of POR to cells in solid tumors. The toxicity of POR to marrow stem cells (CFU-GM and CFU-MK) was not altered by DIC. Pretreatment of mice with DIC therefore produced a small improvement in the therapeutic ratio.
- Published
- 1989
26. Further clinical trials with porfiromycin (NSC-56410) (large intermittent doses).
- Author
-
Izbicki R, al-Sarraf M, Reed ML, Vaughn CB, and Vaitkevicius VK
- Subjects
- Adenocarcinoma drug therapy, Adult, Anemia chemically induced, Astrocytoma drug therapy, Blood Cell Count, Blood Platelets, Carcinoma drug therapy, Carcinoma, Squamous Cell drug therapy, Child, Clinical Trials as Topic, Cystadenoma drug therapy, Female, Humans, Leukocyte Count, Osteosarcoma drug therapy, Porfiromycin administration & dosage, Porfiromycin adverse effects, Porfiromycin blood, Porfiromycin urine, Rhabdomyosarcoma drug therapy, Neoplasms drug therapy, Porfiromycin therapeutic use
- Published
- 1972
27. Effects of mitomycin C and porfiromycin on exponentially growing and plateau phase cultures.
- Author
-
Rockwell S and Hughes CS
- Subjects
- Aerobiosis, Anaerobiosis, Animals, Cell Division drug effects, Cell Division physiology, Female, Mice, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured pathology, Antibiotics, Antineoplastic pharmacology, Mammary Neoplasms, Experimental pathology, Mitomycin pharmacology, Porfiromycin pharmacology
- Abstract
Laboratory studies and clinical trials are exploring the use of hypoxia-directed cytotoxic agents as adjuncts to radiotherapy. Because hypoxia and the microenvironmental inadequacies associated with hypoxia in solid tumours inhibit cell proliferation, an essential requirement for the successful use of hypoxia-directed drugs in cancer therapy is that these drugs be toxic to quiescent tumour cells, as well as tumour cells progressing rapidly through the cell cycle. The experiments reported here compared the cytotoxicities of mitomycin C and porfiromycin to exponentially growing and plateau phase cultures of EMT6 mouse mammary tumour cells. The proliferative status of the cultures did not influence the cytotoxicity of mitomycin C under either aerobic or hypoxic conditions, or the cytotoxicity of porfiromycin in air. Exponentially growing cultures were slightly more sensitive than plateau phase cultures to porfiromycin in hypoxia, but the difference between the sensitivities of proliferating and quiescent cells was much smaller than the difference between aerobic and hypoxic cells. No evidence for repair of potentially lethal damage was found after treatment with porfiromycin in air or in hypoxia; this is in agreement with previous findings for mitomycin C. Mitomycin C and porfiromycin therefore exhibit the toxicity to quiescent cells needed for effective use as hypoxia-directed drugs for the treatment of solid tumours.
- Published
- 1994
- Full Text
- View/download PDF
28. Isolation and Identification of Metabolites of Porfiromycin Formed in the Presence of a Rat Liver Preparation
- Author
-
Wensheng Lang, Terrence W. Doyle, Qin Wang, John Mao, Bijan Almassian, and Chuansheng Niu
- Subjects
Electrospray ionization ,Metabolite ,Pharmaceutical Science ,High-performance liquid chromatography ,Mass Spectrometry ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Animals ,Cysteine ,Nuclear Magnetic Resonance, Biomolecular ,Biotransformation ,Chromatography, High Pressure Liquid ,Antibiotics, Antineoplastic ,Chromatography ,Chemistry ,Primary metabolite ,Metabolism ,Porfiromycin ,Acetylcysteine ,Rats ,Metabolic pathway ,Liver ,Biochemistry ,Spectrophotometry, Ultraviolet ,Drug metabolism - Abstract
The isolation and identification of the major metabolites of porfiromycin formed in the presence of a rat liver preparation under aerobic conditions were performed with high‐performance liquid chromatography and electrospray ionization mass spectrometry. Porfiromycin was extensively metabolized by the rat liver preparation in an aqueous 0.1 M potassium phosphate buffer (pH 7.4) containing an NADPH generating system at 37°C. A total of eight metabolites was identified as mitosene analogs. Of these, three primary metabolites are 2‐methylamino‐7‐aminomitosene, 1,2‐ cis and 1,2‐ trans ‐1‐hydroxy‐2‐methylamino‐7‐aminomitosene, which are consistent with those previously observed in hypoxia using purified rat liver NADPH‐cytochrome c reductase. Interestingly, 2‐methylamino‐7‐aminomitosene is a reactive metabolite, which undergoes further activation at the C‐10 position by the loss of carbamic acid and then links with the 7‐amino group of the primary metabolites to yield two dimeric adducts. In addition, three phosphate adducts, 10‐decarbamoyl‐2‐methylamino‐7‐aminomitosene‐10‐phosphate, 1,2‐ cis and 1,2‐ trans ‐2‐methylamino‐7‐aminomitosene‐1‐phosphate, were also identified in the incubation system. The configurations of the diastereoisomeric metabolites were determined with 1 HNMR and phosphatase digestion. On the basis of the metabolite profile, we propose in vitro metabolic pathways for porfiromycin. The findings provide direct evidence for understanding the reactive nature and hepatic metabolism of the drug currently in phase III clinical trials. © 2000 Wiley‐Liss, Inc. and the American Pharmaceutical Association J Pharm Sci 89:191–198, 2000
- Published
- 2000
- Full Text
- View/download PDF
29. pH-dependent inactivation of DT-diaphorase by mitomycin C and porfiromycin.
- Author
-
Siegel D, Beall H, Kasai M, Arai H, Gibson NW, and Ross D
- Subjects
- Animals, Chromatography, High Pressure Liquid, Electrophoresis, Polyacrylamide Gel, Humans, Hydrogen-Ion Concentration, Kinetics, NAD metabolism, Rats, Rats, Sprague-Dawley, Tumor Cells, Cultured, Mitomycin pharmacology, NAD(P)H Dehydrogenase (Quinone) antagonists & inhibitors, Porfiromycin pharmacology
- Abstract
Mitomycin C and porfiromycin were found to inactivate rat hepatic DT-diaphorase. Inactivation was pH dependent; little inactivation was detected at pH 5.8, but inactivation increased as the pH was raised to 7.8. Inactivation was concentration and time dependent and displayed pseudo-first-order kinetics. Inactivation was NADH dependent, indicating that reductive metabolism was necessary for inhibition. [3H]Mitomycin C was covalently bound to DT-diaphorase during inhibition, and the stoichiometry for inactivation of DT-diaphorase by mitomycin C was approximately 0.8 nmol of mitomycin C bound/nmol of enzyme. A higher molecular mass product (60 kDa) was detected by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western blot analysis of DT-diaphorase preincubated with NADH and mitomycin C at pH 7.8, suggesting that mitomycin C is capable of cross-linking DT-diaphorase. The kinetics of inhibition, requirement for NADH for inhibition, covalent binding of [3H] mitomycin C to DT-diaphorase, and approximate 1:1 stoichiometry suggest that this inactivation process may be mechanism based. Inhibition of DT-diaphorase by mitomycin C and porfiromycin is not limited to a cell-free system and could also be observed in HT-29 cells in culture at pH 7.2. Bioactivation of mitomycin C or porfiromycin by DT-diaphorase is favored at lower pH, whereas at higher pH values enzyme alkylation and inactivation of DT-diaphorase occur. These data suggest that the success of attempts to exploit the elevated DT-diaphorase content of certain human tumors for improved chemotherapeutic response using mitomycin C or porfiromycin will depend on intracellular pH.
- Published
- 1993
30. Determination of pKa values of some prototropic functions in mitomycin and porfiromycin.
- Author
-
Underberg WJ and Lingeman H
- Subjects
- Chemical Phenomena, Chemistry, Physical, Half-Life, Hydrogen-Ion Concentration, Kinetics, Protons, Stereoisomerism, Mitomycins, Porfiromycin
- Published
- 1983
- Full Text
- View/download PDF
31. Correlation between drug uptake and selective toxicity of porfiromycin to hypoxic EMT6 cells.
- Author
-
Keyes SR, Rockwell S, and Sartorelli AC
- Subjects
- Animals, Cell Survival drug effects, DNA metabolism, Mammary Neoplasms, Experimental metabolism, Mice, Porfiromycin pharmacology, Tumor Cells, Cultured drug effects, Mitomycins pharmacokinetics, Oxygen pharmacology, Porfiromycin pharmacokinetics
- Abstract
Mitomycin C and its methylated analogue porfiromycin (Por) have significant potential as adjuncts to regimens presently used for treating solid tumors because of their preferential toxicity to cells existing in an hypoxic environment. An understanding of the factors producing the differential activity of these drugs under aerobic and hypoxic conditions would facilitate the development of new agents of this class. Previous studies have focused on the enzymes that reductively activate the mitomycins and on the interaction of these drugs with DNA; none of these studies has fully explained the differences in cytotoxicity observed under hypoxic and aerobic conditions. The present investigation demonstrates that the rate of Por uptake is directly correlated with cytotoxicity under both aerobic and hypoxic conditions. Uptake of Por into hypoxic cells is more rapid than into aerobic cells at equal drug concentrations. Hypoxic cells also accumulate drug in concentrations well in excess of those in the extracellular medium; this is apparently a reflection of drug sequestration in these cells. This sequestration of Por, which affects the rate and extent of uptake in hypoxic cells, does not take place in aerobic cells. The failure of aerobic cells to sequester drug is evidenced by the very rapid efflux of Por from these cells upon removal of extracellular Por and by the fact that aerobic cells attain a state of equilibrium between the intracellular and extracellular drug concentrations. The findings demonstrate that differences in the uptake and retention of Por are associated with the preferential toxicity of Por to hypoxic cells.
- Published
- 1987
32. Porfiromycin in the management of epidermoid and transitional cell cancer: a phase II study.
- Author
-
Panettiere FJ, Talley RW, Torres J, and Lane M
- Subjects
- Clinical Trials as Topic, Female, Head and Neck Neoplasms drug therapy, Humans, Leukopenia chemically induced, Lung Neoplasms drug therapy, Porfiromycin adverse effects, Thrombocytopenia chemically induced, Ureteral Neoplasms drug therapy, Urinary Bladder Neoplasms drug therapy, Uterine Cervical Neoplasms drug therapy, Vulvar Neoplasms drug therapy, Carcinoma, Squamous Cell drug therapy, Carcinoma, Transitional Cell drug therapy, Porfiromycin therapeutic use
- Abstract
Porifiromycin has been tested in many groups of patients over the past several years (1-7). This report is an analysis of greater than 100 patients with epidermoid and urinary tract transitional cell carcinomas treated with this agent. A review of these data and the available literature on this agent shows that porfiromycin offers definite usefulness in disseminated squamous cell carcinomas of the cervix, and definite but lesser effectiveness in epidermoid carcinomas of the lung, the head and neck region, and other sites. Responsiveness was also demonstrated in transitional cell carcinomas of the urinary tract. Toxicity was tolerable and consisted primarily of myelosuppression and local skin necrosis in sites where extravasation had occurred.
- Published
- 1976
33. Mechanism of transport and intracellular binding of porfiromycin in HCT 116 human colon carcinoma cells.
- Author
-
Pan SS, Johnson R, Gonzalez H, and Thohan V
- Subjects
- Animals, Biological Transport, Biotransformation, Carbon Radioisotopes, Cell Line, Colonic Neoplasms, Erythrocytes metabolism, Humans, In Vitro Techniques, Kinetics, Microsomes, Liver metabolism, Porfiromycin blood, Rats, Mitomycins metabolism, Porfiromycin metabolism, Tumor Cells, Cultured metabolism
- Abstract
The mechanism of uptake and efflux of porfiromycin (PFM) by HCT 116 human colon carcinoma cells or freshly obtained human RBC was investigated. The time course of uptake of radioactivity upon exposure of HCT 116 cells to [14C]PFM showed one fast and one slow phase of linear increase. The initial phase of PFM uptake was not saturable with external drug concentrations from 2 to 100 microM. PFM accumulation was temperature dependent with a temperature coefficient (Q10 24-37 degrees C) of 2.3 +/- 0.3. PFM uptake was not affected either by individual inhibitors such as 1 mM 2,4-dinitrophenol, sodium azide, iodoacetic acid, ouabain, 0.02 mM oligomycin, p-hydroxylmercuribenzoate, 0.2 mM N-ethylmaleimide, or by combinations of inhibitors. PFM uptake did not demonstrate competitive inhibition by unlabeled PFM and mitomycin C. Efflux of cellular radioactivity was not affected by the above mentioned inhibitors or by verapamil, diltiazem, or trifluoperazine. Only aliphatic alcohols accelerated the initial influx rate. The RBC, however, only exhibited the initial fast accumulation of [14C]PFM, and all the 14C accumulated by RBC was exchangeable. These data demonstrate that the uptake and the efflux of PFM in HCT 116 cells and RBC comprise a passive diffusion process.
- Published
- 1989
34. Aspects of the chemical stability of mitomycin and porfiromycin in acidic solution.
- Author
-
Underberg WJ and Lingeman H
- Subjects
- Drug Stability, Hydrogen-Ion Concentration, Kinetics, Phosphates analysis, Solutions, Temperature, Thermodynamics, Mitomycins, Porfiromycin
- Abstract
Aspects of the degradations of mitomycin and porfiromycin were studied. The initial degradation processes of the compounds in an acidic medium were investigated. Influences of pH, buffers, and other additives such as halogenides and dioctyl sodium sulfosuccinate [sodium 1,4-bis(2-ethylhexyl)sulfosuccinate] were studied. The hydrogen ion catalyzes the degradation of both the uncharged and the protonated species. Anions also promote the degradation of the compounds in an acidic medium. Rate constants for all of the catalytic reactions could be determined. From the pH profiles, after correction for buffer influences, accurate pKa values for the aziridine nitrogens could be obtained. The protective influence of the dioctyl sulfosuccinate ion could be explained. From the data obtained a plausible mechanism for the initial acidic degradation reactions was developed.
- Published
- 1983
- Full Text
- View/download PDF
35. Metabolites and DNA adduct formation from flavoenzyme-activated porfiromycin.
- Author
-
Pan SS and Iracki T
- Subjects
- Alkylating Agents, Chemical Phenomena, Chemistry, Chromatography, High Pressure Liquid, Magnetic Resonance Spectroscopy, Mass Spectrometry, Oxidation-Reduction, Spectrum Analysis, Structure-Activity Relationship, DNA, DNA Damage, Mitomycins, Porfiromycin analogs & derivatives
- Abstract
Porfiromycin was reductively metabolized by NADPH cytochrome P-450 reductase and xanthine oxidase under anaerobic conditions. The production of metabolites varied with the pH and the contents of the reaction buffer. In Tris buffer, two major metabolites were produced at pH 7.5 and above, whereas one major metabolite was produced at pH 6.5. The three major metabolites were separated and isolated by HPLC. Identification by californium-252 plasma desorption mass spectrometry showed that the two major metabolites from pH 7.5 were (trans) and (cis)-forms of 7-amino-1-hydroxyl-2-methylaminomitosene and the major metabolite from pH 6.5 was 7-amino-2-methylaminomitosene. All three major metabolites showed substitutions at the C-1 position. DNA was alkylated readily by enzyme-activated porfiromycin. Digestion of porfiromycin-alkylated DNA by DNase, snake venom phosphodiesterase, and alkaline phosphatase resulted in an insoluble nuclease-resistant fraction and a soluble fraction. The nuclease-resistant fraction reflected a high content of cross-linked adducts. Upon HPLC analysis, the solubilized fraction contained two monofunctionally linked porfiromycin adducts and a possibly cross-linked dinucleotide. The major adduct was isolated by HPLC and identified by NMR, as N2-(2'-deoxyguanosyl)-7-amino-2-methylaminomitosene. The N2 position of deoxyguanosine appeared as the major monofunctional alkylating site for DNA alkylation by porfiromycin. Thus, mitomycin C and porfiromycin (which differs from mitomycin C only by the addition of a methyl group to the aziridine nitrogen) share the same enzymatic activating mechanism that leads to the formation of the same types of metabolites and the same specificity of DNA alkylation.
- Published
- 1988
36. ISOLATION AND IDENTIFICATION OF PORFIROMYCIN-DNA ADDUCTS PRODUCED IN LIVING TUMOR CELLS
- Author
-
Hughes, C.S., primary, Tomasz, M., additional, Chowdary, D., additional, Keyes, S.R., additional, Lipman, R., additional, Sartorelli, A.C., additional, and Rockwell, S., additional
- Published
- 1991
- Full Text
- View/download PDF
37. Potentiation of mitomycin C and porfiromycin antitumor activity in solid tumor models by recombinant human interleukin 1 alpha.
- Author
-
Braunschweiger PG, Jones SA, Johnson CS, and Furmanski P
- Subjects
- Animals, Dexamethasone pharmacology, Drug Screening Assays, Antitumor, Drug Synergism, Female, Humans, Interleukin-1 administration & dosage, Interleukin-1 antagonists & inhibitors, Ketoconazole pharmacology, Mice, Mice, Inbred C57BL, Mitomycin administration & dosage, Porfiromycin administration & dosage, Recombinant Proteins therapeutic use, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasms drug therapy
- Abstract
The time- and dose-dependent effects of recombinant human interleukin 1 alpha (IL-1 alpha) on the antitumor activity of mitomycin C (MMC) and porfiromycin (PORF) were studied in RIF-1 and Panc02 solid tumor model systems. IL-1 alpha produced dose-dependent sensitization of clonogenic RIF-1 tumor cells to MMC in vivo. IL-1 alpha chemosensitization was highly schedule dependent, and the most efficacious schedules produced dose-modifying factors of 3.6 and 5.1 for MMC and PORF, respectively. More than additive clonogenic cell kill after IL-1 alpha-chemotherapy combinations reflected increased cellular sensitivity to MMC and PORF. The combinations also produced marked decreases in the yield of viable tumor cells, suggesting that the bioreductive drugs may have also potentiated the microvascular injury and ischemia produced by IL-1 alpha. Dexamethasone inhibited and ketoconazole, an inhibitor of corticosterone biosynthesis, enhanced IL-1 alpha-mediated chemosensitization in these models. IL-1 alpha mediated chemosensitization to MMC, and PORF was also demonstrated by tumor growth inhibition in the RIF-1 model and increased survival of mice in the spontaneously metastasizing Panc02 system. Chemosensitization of bone marrow spleen colony-forming units was not seen. IL-1 alpha (1000 units/ml) had no effect on MMC and PORF cytotoxicity in RIF-1 and PORF cell lines in vitro. The results indicate that the tumor-specific IL-1 alpha-induced pathophysiologies can sensitize solid tumors to agents which are preferentially activated, retained, and cytotoxic to cells under hypoxic conditions. Our results suggest that strategies combining bioreductively activated hypoxic cell cytotoxins and biological agents might offer efficacious alternatives or adjuvants to conventional combination approaches.
- Published
- 1991
38. Isolation, identification, and assay of [3H]-porfiromycin adducts of EMT6 mouse mammary tumor cell DNA: effects of hypoxia and dicumarol on adduct patterns.
- Author
-
Tomasz M, Hughes CS, Chowdary D, Keyes SR, Lipman R, Sartorelli AC, and Rockwell S
- Subjects
- Alkylation, Animals, Chemical Phenomena, Chemistry, Chromatography, High Pressure Liquid, Dicumarol pharmacology, Mice, Oxygen, Spectrophotometry, Ultraviolet, Tritium, Tumor Cells, Cultured, DNA, Neoplasm analysis, Mammary Neoplasms, Experimental genetics, Porfiromycin analysis
- Abstract
[3H]-(N-la-methyl) Porfiromycin (POR) was employed to detect and identify the radiolabeled mono- and bis-adducts formed in living EMT6 mouse mammary tumor cells under different conditions. To provide authentic standard adducts, calf-thymus DNA was treated with POR under reductive activation, then digested to nucleosides and POR-nucleoside adducts. The three major adducts formed were isolated by HPLC and authenticated. Two were mono-adducts, composed of deoxyguanosine linked at its N2-position to C-1 of POR and of 10-decarbamoyl POR. The third was a bis-adduct, in which POR was crosslinked to two deoxyguanosines at their N2-positions. DNA from [3H]-POR treated EMT6 cells was digested an analyzed by HPLC. DNA-associated label was located in thymidine and in two mono-adducts and one bis-adduct identical to those described above. Label in thymidine resulted from N-demethylation of POR and reincorporation of label into new thymidylate residues. Adducts were formed more abundantly in hypoxia than in air. In addition, the mono-adduct to crosslink ratios were different, approximately 1:1 and 2:1 for hypoxic and aerobic cells, respectively. The different patterns of alkylation in air and hypoxia may be related to the greater toxicity of POR in hypoxia. When cells were treated simultaneously with POR and dicumarol, adduct levels were lower, and a new, unknown adduct was observed primarily under hypoxia; these changes may be related to the altered toxicity of POR in the presence of dicumarol. The HPLC assay detected simultaneously the full array of stable mono- and bis-adducts in DNA with good sensitivity (greater than or equal to 2 x 10(6) adducts/nucleotide) and excellent reproducibility. This assay should be generally applicable to all cells and tissues when MC or POR with high specific radioactivity can be employed.
- Published
- 1991
- Full Text
- View/download PDF
39. Studies on the mechanism of resistance to mitomycin C and porfiromycin in a human cell strain derived from a cancer-prone individual.
- Author
-
Marshall RS, Paterson MC, and Rauth AM
- Subjects
- Cell Line drug effects, Cell Line metabolism, Cell Survival drug effects, Child, Female, Glutathione analysis, Humans, Kinetics, Mitomycin, NAD(P)H Dehydrogenase (Quinone), Oxygen, Precipitin Tests, Quinone Reductases analysis, Quinone Reductases genetics, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured metabolism, Drug Resistance genetics, Mitomycins pharmacology, Neoplasms genetics, Porfiromycin pharmacology
- Abstract
The mechanism of aerobic resistance to the quinone-containing anti-tumour agents mitomycin C (MMC) and porfiromycin (PM) has been investigated using non-transformed human cells. One of the cell strains used (3437T) was derived from an afflicted member of a cancer-prone family. This cell strain had been shown previously to be six times more resistant to the cytotoxic effects of these agents under aerobic but not hypoxic conditions when compared to a cell strain derived from an unrelated, normal donor (GM38). Differences could not be detected in the ability of cell sonicates prepared from either cell strain to produce alkylating species under aerobic conditions using a 4-(p-nitrobenzyl)pyridine assay. However, using 3H-labelled PM to monitor rapid drug uptake and subsequent accumulation due to drug metabolism, results were obtained indicating that the resistant cell strain (3437T) was deficient in an enzymatic pathway capable of metabolizing these compounds under aerobic but not hypoxic conditions. Dicumarol, an inhibitor of the quinone reductase DT-diaphorase (EC 1.6.99.2), decreased aerobic drug accumulation and cytotoxicity in the control cell strain, but did not alter the lack of accumulation noted in the resistant cell strain. Under hypoxic conditions, dicumarol increased cytotoxicity and drug accumulation in both cell strains. The mechanism of this enhanced cytotoxicity remains unclear. These results suggested that the resistant cells were deficient in the enzyme DT-diaphorase, a potential activator of PM. Enzymatic assays confirmed this and revealed no alterations in cytochrome P450 reductase (EC 1.6.2.4) activity or glutathione content. No protein characteristic of DT-diaphorase was detected in the resistant cell strain using a polyclonal rabbit-anti-rat antibody raised against this enzyme. Southern blot analysis using a rat DT-diaphorase cDNA probe demonstrated differences between the normal and resistant cell strains in the restriction fragment patterns. The present results are consistent with the hypothesis that decreased DT-diaphorase levels are causally associated with PM and MMC resistance in these cells under aerobic exposure conditions.
- Published
- 1991
- Full Text
- View/download PDF
40. Distribution of porfiromycin in EMT6 solid tumors and normal tissues of BALB/c mice.
- Author
-
Keyes SR, Rockwell S, Kennedy KA, and Sartorelli AC
- Subjects
- Animals, Bone Marrow metabolism, Brain metabolism, Injections, Intraperitoneal, Intestine, Small metabolism, Kidney metabolism, Liver metabolism, Lung metabolism, Mammary Neoplasms, Experimental drug therapy, Mice, Mice, Inbred BALB C, Myocardium metabolism, Tissue Distribution, Urinary Bladder metabolism, Mammary Neoplasms, Experimental metabolism, Porfiromycin pharmacokinetics
- Abstract
The distribution of porfiromycin was studied in BALB/c mice bearing EMT6 mammary tumors. The levels of 3H in blood and most tissues peaked approximately 15 minutes after intraperitoneal injection of [3H]porfiromycin. The levels of radioactivity present in most of the tissues and in the tumors were similar at 4 hours and 24 hours after administration. Most of the normal tissues showed uniform, low grain densities when analyzed by autoradiography; the liver and the small intestine had the highest labeling densities. Only kidney, bladder, and tumor showed differential distributions of grains from [3H]porfiromycin. In the kidney, higher grain counts were found in cortex than in medullary regions; grains were uniformly distributed within each region. In the bladder, the highest labeling densities were found in regions near the lumen. Tumor regions that had some necrotic features or regions of necrosis that included some viable cells showed higher labeling intensities than healthy-looking tumor regions, probably because the abnormal microenvironments in these regions led to increased rates of activation of porfiromycin to electrophilic species. These findings show that porfiromycin can reach and be activated in tumor regions containing cells resistant to many chemotherapeutic agents and to x rays. The results also support the concept that agents such as porfiromycin can target cells in specific microenvironmental subpopulations of solid tumors.
- Published
- 1991
- Full Text
- View/download PDF
41. Effect of deficiencies in DNA repair on the toxicity of mitomycin C and porfiromycin to CHO cells under aerobic and hypoxic conditions.
- Author
-
Hughes CS, Irvin CG, and Rockwell S
- Subjects
- Aerobiosis, Animals, Cell Hypoxia, Cell Line, Cell Survival drug effects, Cricetinae, Dose-Response Relationship, Drug, Female, In Vitro Techniques, Mitomycin, Time Factors, Antineoplastic Agents toxicity, DNA Repair, Mitomycins toxicity, Porfiromycin toxicity
- Abstract
A wild type Chinese hamster cell line (AA8) and three repair-deficient sublines of AA8 (EM9, UV4, and UV5) were used to study the nature of the cytotoxic lesions produced by the bioreductive alkylating agents mitomycin C and porfiromycin under aerobic and hypoxic conditions. The sensitivities of the repair-deficient sublines to the drugs varied markedly: EM9 was similar to AA8, whereas UV4 was exquisitely sensitive and UV5 was of intermediate sensitivity. Moreover, both the relative toxicities of the two drugs and the relative toxicities of each drug under aerobic and hypoxic conditions varied for the different cell lines. These data suggest that there are differences in the spectra of toxic lesions produced by mitomycin and porfiromycin and that there are differences in the lesions produced by these drugs under aerobic and hypoxic conditions.
- Published
- 1991
42. PORFIROMYCIN.
- Author
-
DUVALL LR
- Subjects
- Animals, Dogs, Mice, Rats, Anti-Bacterial Agents, Antibiotics, Antitubercular, Antineoplastic Agents, Chemical Phenomena, Chemistry, Dermatologic Agents, Haplorhini, Mitomycin, Mitomycins, Pharmacology, Porfiromycin, Research, Toxicology
- Published
- 1963
43. Survival of hematopoietic and leukemic colony-forming cells in vivo after administration of mitomycin C or porfiromycin.
- Author
-
Razek A, Valeriote F, and Vietti T
- Subjects
- Animals, Cell Division, Cell Survival drug effects, Clone Cells, Dose-Response Relationship, Drug, Female, Male, Mice, Mice, Inbred AKR, Mitomycins therapeutic use, Porfiromycin therapeutic use, Rats, Time Factors, Hematopoietic Stem Cells drug effects, Leukemia, Experimental drug therapy, Mitomycins pharmacology, Porfiromycin pharmacology
- Published
- 1973
- Full Text
- View/download PDF
44. The identity of porfiromycin and methyl mitomycin.
- Author
-
WAKAKI S, HARADA Y, UZU K, WHITFIELD GB, WILSON AN, KALOWSKY A, STAPLEY EO, WOLF FJ, and WILLIAMS DE
- Subjects
- Anti-Bacterial Agents chemistry, Antibiotics, Antitubercular, Antineoplastic Agents chemistry, Mitomycins, Porfiromycin
- Published
- 1962
45. PREDICTION OF STABILITY IN PAHRMACEUTICAL PREPARATIONS. 13. STABILITY, SPECTROPHOTOMETRIC , AND BIOLOGICAL ASSAY OF THE ANTIBIOTIC PORFIROMYCIN IN PHARMACEUTICALLY USEFUL PH RANGES.
- Author
-
GARRETT ER and SCHROEDER W
- Subjects
- Buffers, Anti-Bacterial Agents, Antibiotics, Antitubercular, Antineoplastic Agents, Biological Assay, Chemistry, Pharmaceutical, Chromatography, Hydrogen-Ion Concentration, Porfiromycin, Research, Spectrophotometry
- Published
- 1964
- Full Text
- View/download PDF
46. SYNERGISM OF THE ANTINEOPLASTIC ACTIVITY OF CYTOSINE ARABINOSIDE BY PORFIROMYCIN.
- Author
-
EVANS JS, BOSTWICK L, and MENGEL GD
- Subjects
- Animals, Mice, Anti-Bacterial Agents, Antineoplastic Agents, Body Weight, Cytarabine, Cytosine, Leukemia, Leukemia L1210, Leukemia, Experimental, Mitomycin, Mitomycins, Neoplasms therapy, Neoplasms, Experimental, Nucleosides, Pharmacology, Porfiromycin, Research, Survival
- Published
- 1964
- Full Text
- View/download PDF
47. Studies on the antimicrobial action of porfiromycin.
- Author
-
PITTILLO RF and QUINNELLY BG
- Subjects
- Anti-Bacterial Agents pharmacology, Anti-Infective Agents, Antibiotics, Antitubercular, Antineoplastic Agents pharmacology, Dermatologic Agents, Porfiromycin
- Published
- 1962
48. Porfiromycin antitumor and toxicopathologic studies.
- Author
-
EVANS JS, MUSSER EA, and GRAY JE
- Subjects
- Anti-Bacterial Agents pharmacology, Antibiotics, Antitubercular, Antineoplastic Agents pharmacology, Dermatologic Agents, Porfiromycin
- Published
- 1961
49. THE PHYSICAL CHEMICAL CHARACTERIZATION OF THE PRODUCTS, EQUILIBRIA, AND KINETICS OF THE COMPLEX TRANSFORMATIONS OF THE ANTIBIOTIC PORFIROMYCIN.
- Author
-
GARRETT ER
- Subjects
- Kinetics, Anti-Bacterial Agents, Antibiotics, Antitubercular, Chemical Phenomena, Chemistry, Dermatologic Agents, Pharmacology, Porfiromycin, Research, Spectrum Analysis
- Published
- 1963
- Full Text
- View/download PDF
50. Addition of a hypoxic cell selective cytotoxic agent (mitomycin C or porfiromycin) to Fluosol-DA/carbogen/radiation.
- Author
-
Holden SA, Herman TS, and Teicher BA
- Subjects
- Animals, Cell Hypoxia drug effects, Cell Hypoxia radiation effects, Dose-Response Relationship, Radiation, Drug Combinations pharmacology, Hydroxyethyl Starch Derivatives, Male, Mice, Mitomycin, Tumor Cells, Cultured, Carbon Dioxide, Fluorocarbons pharmacology, Mitomycins pharmacology, Neoplasms, Experimental pathology, Oxygen, Porfiromycin pharmacology, Radiation-Sensitizing Agents
- Abstract
In an effort to develop effective combination treatments for use with radiation against solid tumors, the cytotoxic effects of the addition of mitomycin C or porfiromycin on treatment with Fluosol-DA/carbogen (95% O2/5% CO2) breathing and radiation in the FSaIIC tumor system were studied. In vitro mitomycin C and porfiromycin were both preferentially cytotoxic toward hypoxic FSaIIC cells. After in vivo exposure, however, the cytotoxicity of mitomycin C toward single cell tumor suspensions obtained from whole tumors was exponential over the dose range studied, but for porfiromycin a plateau in cell killing was observed. With Fluosol-DA/carbogen breathing and single dose radiation, addition of either mitomycin C or porfiromycin increased the tumor cell kill achieved at 5 Gy by approximately 1.2 and 1.0 logs, respectively. Less effect was seen with addition of the drugs at the 10 and 15 Gy radiation doses. In tumor growth delay experiments, the addition of either mitomycin C or porfiromycin to Fluosol-DA/carbogen breathing and radiation resulted in primarily an additive increase in tumor growth delay. The survival of Hoechst 33342 dye-selected tumor cell subpopulations indicated that Fluosol-DA/carbogen breathing increased the cytotoxicity of radiation (10 Gy) more in the bright cell subpopulation (4-fold) than in the dim cell subpopulation (2-fold) resulting in an overall 4-fold sparing of the dim subpopulation. Mitomycin C and porfiromycin were both more toxic toward the dim cell subpopulations. Addition of mitomycin C or porfiromycin to Fluosol-DA/carbogen breathing and radiation (10 Gy) resulted in a primarily additive effect of the drugs and radiation killing in both tumor cell subpopulations. Thus, with mitomycin C/Fluosol-DA/carbogen and radiation there was a 2-fold sparing of dim cells and with porfiromycin in the combined treatment a 1.6-fold sparing of the dim cell population. Our results indicate that treatment strategies directed against both oxic and hypoxic tumor subpopulations can markedly increase the tumor cell kill achieved by radiation.
- Published
- 1990
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.