Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susan R. Keyes is active.

Publication


Featured researches published by Susan R. Keyes.


Radiation Research | 1988

Preclinical Studies of Porfiromycin as an Adjunct to Radiotherapy

Sara Rockwell; Susan R. Keyes; Alan C. Sartorelli

The bioreductive alkylating agent porfiromycin (POR) is more toxic to EMT6 cells that are hypoxic at the time of treatment than to aerobic cells. The toxicity of POR to hypoxic EMT6 cells in vitro was similar to that of mitomycin C (MC): the aerobic toxicity of POR was considerably less than that of MC. Treatment of cells in vitro with POR before and during irradiation did not sensitize either hypoxic or aerobic cells to X rays; instead, only additive cytotoxicity was produced. In contrast, treatment of solid EMT6 tumors in vivo with POR plus radiation produced supra-additive cytotoxicity, as assessed by analyses of the complete dose-response curves for the killing of tumor cells by radiation alone or by POR alone. The supra-additivity of the combination regimens appeared to reflect the preferential killing by each agent of those tumor cells which were in an environment conferring resistance to the other agent. In contrast, combinations of POR and X rays produced only additive cytotoxicities to marrow CFU-GM. Supra-additive antineoplastic effects were obtained at doses of POR which produced little hematologic or other host toxicity. The complementary cytotoxicities of radiation and POR to cells in different microenvironments in solid tumors and the absence of a similar effect in normal tissue make optimized regimens combining radiotherapy and POR unusually promising for the treatment of solid tumors.


Advances in Enzyme Regulation | 1985

Chemotherapeutic attack of hypoxic tumor cells by the bioreductive alkylating agent mitomycin C

Susan R. Keyes; David C. Heimbrook; Paula M. Fracasso; Sara Rockwell; Stephen G. Sligar; Alan C. Sartorelli

Since the cure of solid tumors is limited by the presence of cells with low oxygen contents, we have approached the development of treatment regimens and of new drugs for these tumors by investigating agents which are preferentially bioactivated under hypoxia. Major emphasis has been directed at studying the mode of action of the mitomycin antibiotics, as bioreductive alkylating agents. Using primarily the EMT6 mouse mammary carcinoma as a solid tumor model, we have found that mitomycin C and porfiromycin are preferentially toxic to cells with low oxygen contents. The mitomycin analog BMY-25282 is more toxic to hypoxic cells than are mitomycin C and porfiromycin; however, unlike these antibiotics, BMY-25282 is preferentially toxic to well-oxygenated cells. With these three mitomycins, we have observed a correlation between cytotoxicity to hypoxic cells, the rate of generation of reactive products, and the redox potentials of the drugs. Investigations of the enzymes in EMT6 cells that could possibly activate mitomycin C have revealed that cytochrome P-450 and xanthine oxidase are not present in measurable quantities and therefore are not responsible for activation of mitomycin C. Activities representative of NADPH-cytochrome c reductase and DT-diaphorase are present in these neoplastic cells. Comparison of these enzymatic activities in EMT6, CHO, and V79 cells with the rate of generation of reactive products under hypoxia shows a direct correlation between these two parameters, but there is no quantitative correlation between these two parameters and the amount of cytotoxicity. Use of purified NADPH-cytochrome c reductase and inhibitors of this enzyme demonstrated that NADPH-cytochrome c reductase can activate mitomycin C, but that it is probably not the only enzyme participating in this bioactivation in EMT6 cells. The DT-diaphorase inhibitor dicoumarol was employed to show that this enzyme is not involved in the activation of mitomycin C to a cytotoxic agent. Instead, DT-diaphorase appears to metabolize mitomycin C to a nontoxic product. This property has been exploited to develop a new treatment regimen for solid tumors. Using X-rays to eliminate well oxygenated cells of a solid tumor implant of the EMT6 carcinoma, we have found that the combination of dicoumarol plus mitomycin C is more toxic to hypoxic tumor cells in vivo than mitomycin C alone. Furthermore, knowledge of the biochemical mechanism of mitomycin C activation permits a prediction of which tumors can best be treated with this combination of drugs by measuring enzymatic activities in biopsy specimens.


Cancer Chemotherapy and Pharmacology | 1989

Modulation of the antineoplastic efficacy of mitomycin C by dicoumarol in vivo.

Sara Rockwell; Susan R. Keyes; Alan C. Sartorelli

SummaryDicoumarol (DIC) modulates the intracellular metabolism of mitomycin C (MC) in vitro, increasing the toxicity of MC to hypoxic EMT6 cells and decreasing its toxicity to aerobic cells. The present experiments asessed whether DIC could be used to increase the therapeutic ratio attainable in vivo when MC was used as an adjunct to radiotherapy. Experiments with transplanted EMT6 tumors in mice showed that DIC increased the toxicity of MC to hypoxic tumor cells and increased the antineoplastic efficacy of regimens combining MC with radiation. DIC did not increase the hematologic toxicity of MC, and pretreatment with DIC plus MC did not augment radiation-induced skin reactions. The increase in antineoplastic effect was therefore obtained without a concomitant increase in normal tissue toxicities, and therapeutic gain was obtained.


European Journal of Cancer and Clinical Oncology | 1987

The effects of adriamycin on intracellular calcium concentrations of L1210 murine leukemia cell

Susan R. Keyes; John Hickman; Alan C. Sartorelli

Changes in the concentration of intracellular Ca2+ may be an important component of the mechanism of adriamycin toxicity to tumor cells. Adriamycin interacts with the plasma membrane, a phenomenon which may lead to a direct modulation of Ca2+ transport proteins or, since the drug is a quinone, may lead to indirect changes in Ca2+ homeostasis induced by oxidative stress to the cell. The calcium content of L1210 murine leukemia cells treated with adriamycin for up to 6 hr was estimated using the cell-impermeant dye arsenazo-III. Pools of intracellular Ca2+ were released to the extracellular compartment, where they reacted with the dye by sequential treatment of the cells with m-fluorocarbonylcyanidediphenylhydrazone (FCCP) and the Ca2+-ionophore A23187. Pretreatment of L1210 cells with ruthenium red (5 microM) selectively decreased the FCCP-releasable Ca2+ pool, which suggested it was mitochondrial in origin. Continuous exposure of L1210 murine leukemia cells in vitro to 5 or 10 microM adriamycin for 2 hr did not produce any change in the intracellular concentration of releasable Ca2+; at 4 hr, however, the total releasable pool of Ca2+ rose by 29% and 46% for 5 and 10 microM adriamycin respectively. This increase was seen predominantly in the mitochondrial pool. Exposure of L1210 cells to the quinone, menadione, also increased the releasable pools of cellular Ca2+ but like adriamycin, only after an incubation period of 4 hr. These results contrasted with a rapid decrease in mitochondrial Ca2+ concentration produced by a short (5 min) exposure to 500 microM t-butylhydroperoxide, a generator of free radicals. After treatment with 8 mM lidocaine, a membrane fluidizing agent, there was a rapid fall in extramitochondrial Ca2+. These findings suggest that changes in L1210 Ca2+ homeostasis induced by adriamycin and menadione are late, and possibly common, events of quinone toxicity to L1210 cells, adriamycin does not have an immediate effect on Ca2+ ion transport produced by the direct interaction of the antibiotic with the plasma membrane, and oxidative stress induced by redox-active quinones may not be important for the induction of toxicity in neoplastic cells.


Cancer communications | 1991

Isolation, identification, and assay of [3H]-porfiromycin adducts of EMT6 mouse mammary tumor cell DNA: effects of hypoxia and dicumarol on adduct patterns.

Maria Tomasz; Christine S. Hughes; Dondapati Chowdary; Susan R. Keyes; Roselyn Lipman; Alan C. Sartorelli; Sara Rockwell

[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.


Leukemia Research | 1987

Induction of the differentiation of HL-60 promyelocytic leukemia cells by palmitoleic and myristoleic acids

Diane M. Jenis; Susan R. Keyes; Alan C. Sartorelli

Exposure of HL-60 promyelocytic leukemia cells to palmitoleic or myristoleic acids for 6 days produced both functional and morphological granulocytic maturation. Considerably less or no induction of differentiation occurred with a variety of other fatty acids. Combinations of fatty acids with the granulocytic inducer of maturation, DMSO, did not significantly increase the degree of differentiation of HL-60 cells over that produced by the fatty acids alone. A series of HL-60 cell clones were isolated which differed in sensitivity to the differentiation inducing activities of palmitoleic acid, myristoleic acid, and DMSO. These findings imply that myristoleic acid and palmitoleic acid act to initiate the maturation process by events that are distinct from those produced by DMSO. The capacity of myristoleic and palmitoleic acids to induce leukemic cell differentiation is discussed with respect to protein acylation by fatty acids.


Cancer communications | 1991

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

Sara Rockwell; Susan R. Keyes; Regina Loomis; Marianne Kelley; Dolatrai M. Vyas; Henry Wong; Terrence W. Doyle; Alan C. Sartorelli

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.


Experimental Gerontology | 1993

Porfiromycin as an adjunct to radiotherapy in young and old mice

Sara Rockwell; Christine S. Hughes; Susan R. Keyes; Alan C. Sartorelli; Katherine A. Kennedy

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.


Advances in Enzyme Regulation | 1995

Preferential kill of hypoxic EMT6 mammary tumor cells by the bioreductive alkylating agent porfiromycin

Alan C. Sartorelli; Michael F. Belcourt; Susan R. Keyes; Chris A. Pritsos; Sara Rockwell

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.


Cancer Research | 1984

Role of NADPH: cytochrome c reductase and DT-diaphorase in the biotransformation of mitomycin C

Susan R. Keyes; Paula M. Fracasso; David C. Heimbrook; Sara Rockwell; Stephen G. Sligar; Alan C. Sartorelli

Collaboration


Dive into the Susan R. Keyes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katherine A. Kennedy

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Maria Tomasz

City University of New York

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roselyn Lipman

City University of New York

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge