Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Semin Hematol ; 58(1): 35-44, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33509441

RESUMEN

One mechanism by which lymphoid malignancies resist standard apoptosis-intending (cytotoxic) treatments is genetic attenuation of the p53/p16-CDKN2A apoptosis axis. Depletion of the epigenetic protein DNA methyltransferase 1 (DNMT1) using the deoxycytidine analog decitabine is a validated approach to cytoreduce malignancy independent of p53/p16. In vivo decitabine activity, however, is restricted by rapid catabolism by cytidine deaminase (CDA). We, therefore, combined decitabine with the CDA-inhibitor tetrahydrouridine and conducted a pilot clinical trial in patients with relapsed lymphoid malignancies: the doses of tetrahydrouridine/decitabine used (∼10/0.2 mg/kg orally (PO) 2×/week) were selected for the molecular pharmacodynamic objective of non-cytotoxic, S-phase dependent, DNMT1-depletion, guided by previous Phase 1 studies. Patients with relapsed/refractory B- or T-cell malignancies (n = 7) were treated for up to 18 weeks. Neutropenia without concurrent thrombocytopenia is an expected toxicity of DNMT1-depletion and occurred in all patients (Grade 3/4). Subjective and objective clinical improvements occurred in 4 of 7 patients, but these responses were lost upon treatment interruptions and reductions to manage neutropenia. We thus performed parallel experiments in a preclinical in vivo model of lymphoma to identify regimen refinements that might sustain DNMT1-targeting in malignant cells but limit neutropenia. We found that timed-alternation of decitabine with the related molecule 5-azacytidine, and combination with inhibitors of CDA and de novo pyrimidine synthesis could leverage feedback responses of pyrimidine metabolism to substantially increase lymphoma cytoreduction but with less neutropenia. In sum, regimen innovations beyond incorporation of a CDA-inhibitor are needed to sustain decitabine DNMT1-targeting and efficacy against chemo-resistant lymphoid malignancy. Such potential solutions were explored in preclinical in vivo studies.


Asunto(s)
Antimetabolitos Antineoplásicos , Tetrahidrouridina , Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/farmacología , Azacitidina/uso terapéutico , Decitabina/farmacología , Decitabina/uso terapéutico , Epigénesis Genética , Humanos , Linfoma/tratamiento farmacológico , Proyectos Piloto , Tetrahidrouridina/farmacología , Tetrahidrouridina/uso terapéutico
2.
J Neurooncol ; 126(2): 225-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26518542

RESUMEN

Chemotherapies active in preclinical studies frequently fail in the clinic due to lack of efficacy, which limits progress for rare cancers since only small numbers of patients are available for clinical trials. Thus, a preclinical drug development pipeline was developed to prioritize potentially active regimens for pediatric brain tumors spanning from in vitro drug screening, through intracranial and intra-tumoral pharmacokinetics to in vivo efficacy studies. Here, as an example of the pipeline, data are presented for the combination of 5-fluoro-2'-deoxycytidine and tetrahydrouridine in three pediatric brain tumor models. The in vitro activity of nine novel therapies was tested against tumor spheres derived from faithful mouse models of Group 3 medulloblastoma, ependymoma, and choroid plexus carcinoma. Agents with the greatest in vitro potency were then subjected to a comprehensive series of in vivo pharmacokinetic (PK) and pharmacodynamic (PD) studies culminating in preclinical efficacy trials in mice harboring brain tumors. The nucleoside analog 5-fluoro-2'-deoxycytidine (FdCyd) markedly reduced the proliferation in vitro of all three brain tumor cell types at nanomolar concentrations. Detailed intracranial PK studies confirmed that systemically administered FdCyd exceeded concentrations in brain tumors necessary to inhibit tumor cell proliferation, but no tumor displayed a significant in vivo therapeutic response. Despite promising in vitro activity and in vivo PK properties, FdCyd is unlikely to be an effective treatment of pediatric brain tumors, and therefore was deprioritized for the clinic. Our comprehensive and integrated preclinical drug development pipeline should reduce the attrition of drugs in clinical trials.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Desoxicitidina/análogos & derivados , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Tetrahidrouridina/administración & dosificación , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/sangre , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Relación Dosis-Respuesta a Droga , Epigénesis Genética/efectos de los fármacos , Ratones , Ratones Desnudos , Tetrahidrouridina/sangre , Tetrahidrouridina/farmacocinética , Tetrahidrouridina/uso terapéutico
3.
Oncotarget ; 3(10): 1137-45, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23087155

RESUMEN

We document for the first time that sanctuary in an organ which expresses high levels of the enzyme cytidine deaminase (CDA) is a mechanism of cancer cell resistance to cytidine analogues. This mechanism could explain why historically, cytidine analogues have not been successful chemotherapeutics against hepatotropic cancers, despite efficacy in vitro. Importantly, this mechanism of resistance can be readily reversed, without increasing toxicity to sensitive organs, by combining a cytidine analogue with an inhibitor of cytidine deaminase (tetrahydrouridine). Specifically, CDA rapidly metabolizes cytidine analogues into inactive uridine counterparts. Hence, to determine if sheltering/protection of cancer cells in organs which express high levels of CDA (e.g., liver) is a mechanism of resistance, we utilized a murine xenotransplant model of myeloid cancer that is sensitive to epigenetic therapeutic effects of the cytidine analogue decitabine in vitro and hepato-tropic in vivo. Treatment of tumor-bearing mice with decitabine (subcutaneous 0.2mg/kg 2X/week) doubled median survival and significantly decreased extra-hepatic tumor burden, but hepatic tumor burden remained substantial, to which the animals eventually succumbed. Combining a clinically-relevant inhibitor of CDA (tetrahydrouridine) with a lower dose of decitabine (subcutaneous 0.1mg/kg 2X/week) markedly decreased liver tumor burden without blood count or bone marrow evidence of myelotoxicity, and with further improvement in survival. In conclusion, sanctuary in a CDA-rich organ is a mechanism by which otherwise susceptible cancer cells can resist the effects of decitabine epigenetic therapy. This protection can be reversed without increasing myelotoxicity by combining tetrahydrouridine with a lower dose of decitabine.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Citidina Desaminasa/metabolismo , Leucemia Mieloide/enzimología , Neoplasias Hepáticas/enzimología , Hígado/enzimología , Animales , Antimetabolitos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Azacitidina/uso terapéutico , Decitabina , Humanos , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Hígado/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Tetrahidrouridina/uso terapéutico , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología
4.
Int J Radiat Oncol Biol Phys ; 51(3): 791-806, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11697326

RESUMEN

PURPOSE: The study's goals were as follows: (1) to extend our past findings with rodent tumors to human tumors in nude mice, (2) to determine if the drug protocol could be simplified so that only CldC and one modulator, tetrahydrouridine (H4U), would be sufficient to obtain efficacy, (3) to determine the levels of deoxycytidine kinase and dCMP deaminase in human tumors, compared to adjacent normal tissue, and (4) to determine the effect of CldC on normal tissue radiation damage to the cervical spinal cord of nude mice. METHODS AND MATERIALS: The five human tumors used were as follows: prostate tumors, PC-3 and H-1579; glioblastoma, SF-295; breast tumor, GI-101; and lung tumor, H-165. The duration of treatment was 3-5 weeks, with drugs administered on Days 1-4 and radiation on Days 3-5 of each week. The biomodulators of CldC were N-(Phosphonacetyl)-L-aspartate (PALA), an inhibitor of aspartyl transcarbamoylase, 5-fluorodeoxycytidine (FdC), resulting in tumor-directed inhibition of thymidylate synthetase, and H4U, an inhibitor of cytidine deaminase. The total dose of focused irradiation of the tumors was usually 45 Gy in 12 fractions. RESULTS: Marked radiosensitization was obtained with CldC and the three modulators. The average days in tumor regrowth delay for X-ray compared to drugs plus X-ray, respectively, were: PC-3 prostate, 42-97; H-1579 prostate, 29-115; glioblastoma, 5-51; breast, 50-80; lung, 32-123. Comparative studies with PC-3 and H-1579 using CldC coadministered with H4U, showed that both PALA and FdC are dispensable, and the protocol can be simplified with equal and possibly heightened efficacy. For example, PC-3 with X-ray and (1) no drugs, (2) CldC plus the three modulators, (3) a high dose of CldC, and (4) escalating doses of CldC resulted in 0/10, 3/9, 5/10, and 6/9 cures, respectively. The tumor regrowth delay data followed a similar pattern. After treating mice only 11/2 weeks with CldC + H4U, 92% of the PC-3 tumor cells were found to possess CldU in their DNA. The great majority of head-and-neck tumors from patient material had markedly higher levels of dC kinase and dCMP deaminase than found in adjacent normal tissue. Physiologic and histologic studies showed that CldC + H4U combined with X-ray, focused on the cervical spinal cord, did not result in damage to that tissue. CONCLUSIONS: 5-CldC coadministered with only H4U is an effective radiosensitizer of human tumors. Ninety-two percent of PC-3 tumor cells have been shown to take up ClUra derived from CldC in their DNA after only 11/2 weeks and 2 weeks of bolus i.p. injections. Enzymatic alterations that make tumors successful have been exploited for a therapeutic advantage. The great electronegativity, coupled with the relatively small Van der Waal radius of the Cl atom, may result in CldC's possessing the dual advantageous properties of FdC on one hand and BrdU and IdU on the other hand. These advantages include autoenhancing the incorporation of CldUTP into DNA by not only overrunning but also inhibiting the formation of competing TTP pools in tumors. A clinical trial is about to begin, with head-and-neck tumors as a first target of CldC radiosensitization.


Asunto(s)
Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Tetrahidrouridina/uso terapéutico , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/radioterapia , Terapia Combinada , DCMP Desaminasa/metabolismo , Desoxicitidina Quinasa/metabolismo , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Masculino , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Dosificación Radioterapéutica , Médula Espinal/efectos de la radiación
5.
J Nucl Med ; 33(8): 1530-4, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1634946

RESUMEN

Recently, there has been much interest in the use of radionuclide conjugated monoclonal antibodies for the treatment of human malignancies. One way to potentially maximize the therapeutic effectiveness of radioimmunotherapy would be to sensitize tumor cells to the radiation dose delivered by the antibody. Since radioimmunotherapy can potentially treat disseminated disease, including micrometastasis, we chose to study a halogenated pyrimidine radiosensitizer, a class of compounds that affect nonhypoxic cells. 5-Iododeoxyuridine, administered with pyrimidine metabolism modulators, increased the therapeutic effectiveness of radioimmunotherapy, resulting in individual cures of human tumors growing in BALB/c nu/nu (nude) mice. 5-Iododeoxyuridine was administered with N-(phosphonacetyl)-L-aspartic acid and 5-fluoro-deoxycytidine plus tetrahydrouridine. This drug treatment was combined with radioimmunotherapy using 131I conjugated to a monoclonal antibody, Mc5. Mc5 binds to a mucin component of the human milk fat globule. This antigen is expressed on the surface of MX-1 cells, the transplantable human tumor used in this study. Tumor-bearing mice treated with both the drug protocol and 131I-Mc5 (540 microCi, 10 microCi/micrograms) showed a regression in average tumor volume. The average tumor volume was reduced below the initial size at treatment for 50 days; two of five cures were obtained. Neither cures nor regressions were observed with either the drug or antibody treatments alone. Our results indicate the potential for increasing the therapeutic effectiveness of radioimmunotherapy of human solid tumors with halogenated pyrimidines.


Asunto(s)
Neoplasias de la Mama/radioterapia , Idoxuridina/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Radioinmunoterapia/métodos , Animales , Ácido Aspártico/administración & dosificación , Ácido Aspártico/análogos & derivados , Ácido Aspártico/uso terapéutico , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Quimioterapia Combinada , Humanos , Idoxuridina/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Ácido Fosfonoacético/administración & dosificación , Ácido Fosfonoacético/análogos & derivados , Ácido Fosfonoacético/uso terapéutico , Tetrahidrouridina/administración & dosificación , Tetrahidrouridina/uso terapéutico , Trasplante Heterólogo
6.
Int J Radiat Oncol Biol Phys ; 19(2): 357-65, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2394614

RESUMEN

Bolus doses of 5-chlorodeoxycytidine (CldC) administered with modulators of pyrimidine metabolism, followed by X-irradiation, resulted in a 2-fold dose increase effect against RIF-1 tumors in C3H mice. Pool size studies of the fate of [14C]-CldC in BDF1 mice bearing Sarcoma-180 tumors, which demonstrated the rapid formation of 5-chlorodeoxycytidylate (CldCMP), and incorporation of CldC as such in RIF-1 tumor DNA, indicate that CldC is a substrate for deoxycytidine kinase, as our past Km studies have shown. Our data indicate that 5-chlorodeoxyuridine triphosphate (CldUTP) accumulates from both the cytidine deaminase-thymidine kinase pathway, as well as from the deoxycytidine kinase-dCMP deaminase pathway, in tumor tissue. As shown in a previous study, tetrahydrouridine (H4U), a potent inhibitor of cytidine deaminase, can effectively inhibit the enzyme in the normal tissues of BDF1 mice. When H4U was administered with the modulators N-(phosphonacetyl)-L-aspartic acid (PALA) and 5-fluorodeoxycytidine (FdC), the levels of CldC-derived RNA and DNA directed metabolites increased in tumor and decreased in normal tissues compared to when CldC was administered alone. These modulators inhibit the de novo pathway of thymidine biosynthesis, lowering thymidine triphosphate (TTP) levels, which compete with CldUTP for incorporation into DNA. 5-Benzylacyclouridine (BAU), an inhibitor of uridine phosphorylase, was also utilized. DNA incorporation studies using C3H mice bearing RIF-1 tumors showed that the extent of incorporation of 5-chlorodeoxyuridine (CldU) into DNA correlates with the levels of cytidine and dCMP deaminases; this is encouraging in view of their high activity in many human malignancies and the low activities in normal tissues, including those undergoing active replication. Up to 3.9% replacement of thymidine by CldU took place in RIF-1 tumors, whereas incorporation into bone marrow was below our limit of detection. CldC did not result in photosensitization under conditions in cell culture in which radiosensitization to X rays was obtained. Thus, the combination of CldC with modulators of its metabolism has potential as a modality of selective radiosensitization for ultimate clinical use in a wider range of tumors than those of the brain.


Asunto(s)
Desoxicitidina/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Sarcoma Experimental/radioterapia , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/uso terapéutico , Terapia Combinada , Citidina Desaminasa/antagonistas & inhibidores , ADN de Neoplasias/metabolismo , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Quimioterapia Combinada , Ratones , Ratones Endogámicos C3H , Ácido Fosfonoacético/análogos & derivados , Ácido Fosfonoacético/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Sarcoma Experimental/tratamiento farmacológico , Sarcoma Experimental/metabolismo , Tetrahidrouridina/uso terapéutico , Uracilo/análogos & derivados , Uracilo/uso terapéutico , Uridina Fosforilasa/antagonistas & inhibidores
7.
Intervirology ; 30 Suppl 1: 12-25, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2540109

RESUMEN

Severe progressive immunodeficiency syndrome can be induced experimentally with a molecularly cloned isolate of feline leukemia virus (FeLV-FAIDS). The resultant disease syndrome is characterized by persistent viremia, lymphopenia, progressive weight loss, persistent diarrhea, enteropathy, and opportunistic infections. The onset of clinical immunodeficiency disease is prefigured by the replication of the FeLV-FAIDS variant virus in bone marrow and other tissues. The FeLV-FAIDS system can be used to evaluate antiviral agents which act on steps in the replication cycle which are conserved among retroviruses (e.g. reverse transcriptase, protease, assembly). The persistence and magnitude of viremia serves as a useful parameter in antiviral studies because it can be easily measured, presages the eventual development of immunodeficiency, and provides a convenient indicator of therapeutic efficacy either in preventing de novo FeLV infection or in reversing or ameliorating established infection. We describe here the evaluation of 2',3'-dideoxycytidine (ddC) against FeLV-FAIDS infection - both in vitro in cell culture assay systems and in vivo in cats administered ddC either via intravenous bolus dosage or via controlled release subcutaneous implants. We found that, although controlled release delivery of ddC inhibited de novo FeLV-FAIDS replication and delayed onset of viremia when therapy was discontinued (after 3 weeks), an equivalent incidence and level of viremia were established rapidly in both ddC-treated and control cats. The FeLV model, therefore, can be used to assess rapidly experimental single agent or combined antiviral therapies for persistent retrovirus infection and disease.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Gatos , Didesoxinucleósidos/uso terapéutico , Modelos Animales de Enfermedad , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Animales , Línea Celular , Efecto Citopatogénico Viral , Preparaciones de Acción Retardada , Didesoxinucleósidos/administración & dosificación , Evaluación Preclínica de Medicamentos , Implantes de Medicamentos , Quimioterapia Combinada , Inyecciones Intravenosas , Inyecciones Subcutáneas , Virus de la Leucemia Felina/efectos de los fármacos , Leucemia Experimental/tratamiento farmacológico , Infecciones por Retroviridae/tratamiento farmacológico , Organismos Libres de Patógenos Específicos , Tetrahidrouridina/uso terapéutico , Viremia/tratamiento farmacológico , Zalcitabina
10.
Bull Cancer ; 66(1): 89-92, 1979.
Artículo en Inglés | MEDLINE | ID: mdl-420952

RESUMEN

Clinical studies of resistance to cytosine arabinoside have not produced agreement as to the specific biochemical lesions responsible for altered sensitivity, although experimental and clinical work supports the concept that a decreased ability to generate ara-CTP must be the ultimate effect of this lesion. 3-deazauridine, an inhibitor of CTP synthetase, was found to enhance ara-CTP production in murine tumor cells, and in the present study, was shown to inhibit deamination of ara-C at both the nucleoside and nucleotide level. Enhanced ara-CTP formation was observed in cells lacking cytidine deaminase (L1 210 and HL60), indicating that 3-deazauridine inhibition of deoxycytidylate deaminase may be important in this drug interaction.


Asunto(s)
Citarabina/metabolismo , 3-Desazauridina/farmacología , Animales , Citarabina/farmacología , Citarabina/uso terapéutico , Citidina Desaminasa/antagonistas & inhibidores , Interacciones Farmacológicas , Resistencia a Medicamentos , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Tetrahidrouridina/farmacología , Tetrahidrouridina/uso terapéutico
11.
Cancer Treat Rep ; 62(7): 1025-32, 1978 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-80268

RESUMEN

The pharmacokinetics of 5-azacytidine (5-azaCR) and tetrahydrouridine (THU) were considered in evaluating the effect of THU on chemotherapy with 5-azaCR in L1210 leukemia mice. The administration of three different dose levels of THU and 5-azaCR ip in either a 6- or 72-hour infusion gave minimal increases in therapeutic effect. At the high-dose combinations (except in the 72-hour infusion), THU appeared to enhance toxicity. Toxicity, however, occurred only after exceeding a theoretic plasma concentration for 5-azaCR of 61 microgram/ml. THU was effective in increasing the excretion of 5-azaCR by sixfold and in altering its urinary metabolites when given simultaneously with or up to 1 hour prior to 5-azaCR.


Asunto(s)
Azacitidina/metabolismo , Leucemia L1210/tratamiento farmacológico , Tetrahidrouridina/metabolismo , Uridina/análogos & derivados , Animales , Azacitidina/administración & dosificación , Azacitidina/uso terapéutico , Quimioterapia Combinada , Femenino , Infusiones Parenterales , Inyecciones Intraperitoneales , Ratones , Tetrahidrouridina/administración & dosificación , Tetrahidrouridina/uso terapéutico
13.
Cancer Treat Rep ; 61(7): 1347-53, 1977 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-589600

RESUMEN

[14C]-tetrahydrouridine (THU), a strong inhibitor of cytidine (CR) deaminase, was, after iv administration, rapidly and quantitatively cleared from the blood with a plasma half-life of about 1 hour. The main pathway of excretion was through the kidneys: most of a dose of 50 mg/kg was excreted within 12 hours and excretion was essentially complete within 48 hours. Oral administration of the same dose revealed absorption of about 10% from the gastrointestinal tract. THU at 10, 25, and 50 mg/kg given 15 minutes before [3H]-cytosine arabinoside (ara-C) at a dose of 0.003 mg/kg produced about a two fold increase in ara-C blood levels at all times measured from 5 minutes to 4 hours, with only slight increases in the half-life of ara-C. A dose-related effect of THU upon the deamination of ara-C was obvious only during the time from 15 minutes to 1 hour after the injection of 3H-ara-C. The inhibitory effect of THU upon CR deaminase was also reflected in a considerably increased ratio of ara-C/uracil arabinoside in the urine.


Asunto(s)
Citarabina/metabolismo , Tetrahidrouridina/metabolismo , Uridina/análogos & derivados , Administración Oral , Disponibilidad Biológica , Citarabina/uso terapéutico , Citidina Desaminasa/antagonistas & inhibidores , Desaminación , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Semivida , Humanos , Inyecciones Intravenosas , Neoplasias/tratamiento farmacológico , Tetrahidrouridina/administración & dosificación , Tetrahidrouridina/uso terapéutico
14.
Cancer Treat Rep ; 61(7): 1355-64, 1977 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-589601

RESUMEN

Thirteen experimental mouse neoplasms were tested by cytidine (CR)-deaminase and deoxycytidine (dCR)-kinase levels. Four neoplasms, Sarcoma T241, Adenocarcinoma E0771, Lewis lung carcinoma (LL), and Sarcoma 180 Japan (S180J), considered to have high deaminase and sufficient dCR-kinase activities, were tested in vivo for combination chemotherapy with cytosine arabinoside (ara-C) and the CR-deaminase inhibitor, tetrahydrouridine (THU). THU did not significantly improve the growth inhibition of ara-C in a wide range of combinations in T241, E0771, LL, and the solid form of S180J, but more than doubled the survival time of the S180J ascites-bearing animals. Toxicity in the form of weight loss and toxic deaths was observed in some but not all groups, especially at high dosages of ara-C and THU. Tissue distribution of [3H]-ara-C and [14C]-THU in T241-bearing mice revealed an accelerated clearance of ara-C-derived radioactivity under the influence of THU in the tumor and five host tissues, but not in the small intestines. With the exception of the small intestines, clearance of THU-derived radioactivity was faster in all tissues studied compared to the clearance of [3H]-ara-C-derived radioactivity. Intracellular CR-deaminase levels were inhibited significantly, ie, dose dependent, in tumor and host kidney after a single ip injection of THU to E0771--bearing mice. In the solid S180J, with or without simultaneous ip administration of THU, [3H]-ara-C was not converted to 5'-di- and tri-phosphates at all. In mice bearing the ascites form of S180J, [3H]-ara-C was extensively converted to ara-C 5'-di- and tri-phosphates. THU increased both overall ara-C-derived radioactivity and the relative amounts of ara-C 5'-di- and tri-phosphates.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Citarabina/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Tetrahidrouridina/uso terapéutico , Uridina/análogos & derivados , Animales , Citarabina/metabolismo , Citidina Desaminasa/antagonistas & inhibidores , Citidina Desaminasa/metabolismo , Desaminación , Evaluación de Medicamentos , Quimioterapia Combinada , Femenino , Ratones , Neoplasias Experimentales/enzimología , Fosfotransferasas/metabolismo , Sarcoma Experimental/tratamiento farmacológico , Tetrahidrouridina/metabolismo , Tritio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA