Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Arch. Soc. Esp. Oftalmol ; 93(2): 97-100, feb. 2018.
Artigo em Espanhol | IBECS | ID: ibc-172586

RESUMO

Caso clínico: Un niño de 7 años con xeroderma pigmentoso presenta una recurrencia de un fibroxantoma atípico conjuntival después de 2 cirugías previas. En esta tercera intervención el paciente es tratado mediante escisión quirúrgica de la tumoración más crioterapia en el lecho. Por el riesgo de recurrencia se asoció en el postoperatorio mitomicina C 0,02% tópica con buen resultado clínico. Discusión: La exéresis quirúrgica con crioterapia y mitomicina C tópica es un tratamiento efectivo en el caso de un fibroxantoma atípico con alto potencial de recurrencia e invasión. Es necesario un seguimiento oftalmológico, así como pediátrico general en estos pacientes y asociar ayudas de apoyo (AU)


Case report: A 7 year-old boy with Xeroderma Pigmentosum (XP) and who presents a recurrent conjunctival atypical fibroxanthoma after two surgeries. This is the third procedure and the patient is treated with a surgical excision of the tumour and cryotherapy at the surgical bed. Due to the risk of recurrence, topical Mitomycin C 0,02% was added at post-operative care achieving a good clinical outcome. Discussion: Surgical exeresis with cryotherapy and topical Mitomycin C is an effective treatment for a case of an atypical fibroxanthoma with a high potential for recurrence and invasion. An ophthalmologic follow-up is required for these patients, as well as general paediatric care and support aids (AU)


Assuntos
Humanos , Masculino , Criança , Xantomatose/diagnóstico por imagem , Xantomatose/terapia , Neoplasias da Túnica Conjuntiva/diagnóstico por imagem , Neoplasias da Túnica Conjuntiva/terapia , Xeroderma Pigmentoso/diagnóstico por imagem , Xeroderma Pigmentoso/terapia , Xantomatose/tratamento farmacológico , Xantomatose/cirurgia , Túnica Conjuntiva/diagnóstico por imagem , Túnica Conjuntiva/patologia , Recidiva Local de Neoplasia/cirurgia , Porfiromicina/uso terapêutico , Mitomicina/uso terapêutico
2.
Arch. Soc. Esp. Oftalmol ; 83(7): 445-448, jul. 2008. ilus
Artigo em Es | IBECS | ID: ibc-66569

RESUMO

Casos clínicos: Mujer de 94 años que consulta por chalazión de 8 meses de evolución. Presenta engrosamiento palpebral con necrosis y madarosis, y una adenopatía preauricular. Mujer de 67 años operada de un carcinoma de glándulas sebáceas que acude por enrojecimiento conjuntival y palpebral. La biomicroscopía manifestaba inflamación del párpado y presencia de una conjuntiva bulbar superior irregular y muy eritematosa con desestructuración de la arquitectura limbar. Discusión: El carcinoma de glándulas sebáceas es un tumor de diagnóstico y manejo difícil ya que puede presentarse de manera parcheada y tiene tendencia a la diseminación pagetoide. Para su diagnóstico se debe realizar biopsia escisional y mapa de biopsias de conjuntiva palpebral y bulbar. El tratamiento posterior dependerá de la extensión del tumor, bien escisión simple, Mitomicina C tópica, radioterapia o exenteración orbitaria


Clinical cases: A 94-year-old woman, who had had a chalazion for a period of 8 months, presented because of thickening of the eyelid with necrosis, madarosis and adenopathy. A 67-year-old woman, previously operated on for a sebaceous carcinoma, presented because of reddening of the conjunctiva and eyelid. Clinical evaluation revealed inflammation of the eyelid and an irregular and erythematous superior bulbar conjunctiva with disruption of the limbal architecture. Discussion: A sebaceous carcinoma is a tumour which is difficult to diagnose and treat, because it can be patchy and has a tendency to pagetoid dissemination. Diagnosis requires a biopsy of the lesion and mapping of biopsies from the conjunctiva of the eyelid and eyebrow. The subsequent treatment depends on the extent of the tumour, and may involve simple cleavage, topical mitomycin C, radiotherapy or exenteration of the eyebrow


Assuntos
Humanos , Feminino , Pessoa de Meia-Idade , Idoso , Neoplasias das Glândulas Sebáceas/complicações , Neoplasias das Glândulas Sebáceas/diagnóstico , Carcinoma/complicações , Carcinoma/diagnóstico , Porfiromicina/uso terapêutico , Blefaroptose/complicações , Conjuntivite/complicações , Conjuntivite/diagnóstico , Blefarite/radioterapia , Biópsia , Tomografia Computadorizada de Emissão
4.
Int J Radiat Oncol Biol Phys ; 61(1): 119-28, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15629602

RESUMO

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.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Mitomicina/uso terapêutico , Análise de Variância , Terapia Combinada , Intervalo Livre de Doença , Feminino , Humanos , Leucopenia/induzido quimicamente , Masculino , Pessoa de Meia-Idade , Porfiromicina/uso terapêutico , Estudos Prospectivos , Dosagem Radioterapêutica , Estatísticas não Paramétricas , Trombocitopenia/induzido quimicamente
5.
J Am Chem Soc ; 126(13): 4281-92, 2004 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-15053618

RESUMO

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.


Assuntos
Antineoplásicos/química , Antineoplásicos/síntese química , Iminas/química , Iminas/síntese química , Porfiromicina/química , Porfiromicina/síntese química , Animais , Antineoplásicos/farmacologia , Hipóxia Celular , Linhagem Celular Tumoral , Adutos de DNA/química , Iminas/farmacologia , Porfiromicina/análogos & derivados , Porfiromicina/farmacologia
8.
Drug Metab Dispos ; 28(8): 899-904, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10901698

RESUMO

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.


Assuntos
Porfiromicina/urina , Compostos Radiofarmacêuticos/urina , Animais , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/urina , Cromatografia Líquida de Alta Pressão , Cães , Humanos , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Porfiromicina/química , Porfiromicina/isolamento & purificação , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/isolamento & purificação , Trítio
9.
J Pharm Sci ; 89(2): 191-8, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10688748

RESUMO

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.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Fígado/metabolismo , Porfiromicina/isolamento & purificação , Porfiromicina/metabolismo , Acetilcisteína/metabolismo , Animais , Antibióticos Antineoplásicos/isolamento & purificação , Biotransformação , Cromatografia Líquida de Alta Pressão , Cisteína/metabolismo , Fígado/química , Espectrometria de Massas , Ressonância Magnética Nuclear Biomolecular , Porfiromicina/farmacocinética , Ratos , Ratos Sprague-Dawley , Espectrofotometria Ultravioleta
10.
Proc Natl Acad Sci U S A ; 96(18): 10489-94, 1999 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10468636

RESUMO

The mitomycin C-resistance gene, mcrA, of Streptomyces lavendulae produces MCRA, a protein that protects this microorganism from its own antibiotic, the antitumor drug mitomycin C. Expression of the bacterial mcrA gene in mammalian Chinese hamster ovary cells causes profound resistance to mitomycin C and to its structurally related analog porfiromycin under aerobic conditions but produces little change in drug sensitivity under hypoxia. The mitomycins are prodrugs that are enzymatically reduced and activated intracellularly, producing cytotoxic semiquinone anion radical and hydroquinone reduction intermediates. In vitro, MCRA protects DNA from cross-linking by the hydroquinone reduction intermediate of these mitomycins by oxidizing the hydroquinone back to the parent molecule; thus, MCRA acts as a hydroquinone oxidase. These findings suggest potential therapeutic applications for MCRA in the treatment of cancer with the mitomycins and imply that intrinsic or selected mitomycin C resistance in mammalian cells may not be due solely to decreased bioactivation, as has been hypothesized previously, but instead could involve an MCRA-like mechanism.


Assuntos
Proteínas de Bactérias/metabolismo , Mitomicina/farmacologia , Oxirredutases , Aerobiose , Animais , Proteínas de Bactérias/genética , Biotransformação , Células CHO , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Clonagem Molecular , Cricetinae , Resistência a Medicamentos , Mitomicina/farmacocinética , Porfiromicina/farmacologia , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Proteínas Recombinantes/metabolismo , Streptomyces/efeitos dos fármacos , Streptomyces/genética , Streptomyces/fisiologia , Transfecção
11.
Adv Enzyme Regul ; 38: 111-33, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9762350

RESUMO

We have directly demonstrated the involvement of human NADPH: cytochrome c (P-450) reductase in the aerobic/hypoxic differential toxicity of mitomycin C and porfiromycin in living cells by varying only this enzyme in a transfected cell line. In the same manner, we have implicated rat DT-diaphorase in the aerobic and hypoxic activation of mitomycin C, but found only a minor role for this enzyme in the aerobic activation of porfiromycin. DT-Diaphorase does not cause the production of an aerobic/hypoxic differential toxicity by mitomycin C, but rather activates this agent through an oxygen insensitive pathway. The evidence suggests that DT-diaphorase activates mitomycin C more effectively than porfiromycin, with porfiromycin being preferentially activated through a one-electron reductive pathway. The therapeutic potential of mitomycin antibiotics in the treatment of cancer can be envisioned to be enhanced for those tumors containing elevated levels of the bioreductive enzymes. However, cytogenetic heterogeneity within the tumor cell population and the various environmental factors which impact on bioreductive enzyme function, including pH and oxygen tension, may subvert this approach. Moreover, if high tumor levels of a drug activating enzyme reflect high levels in the normal tissues of the patient, normal tissue damage may also be enhanced with possibly no improvement in the therapeutic ratio. Approaches utilizing gene therapy, whereby a specific bioreductive catalyst is introduced into the tumor cell population via a targeting vehicle to activate a particular prodrug, may be more effective in that not only will the prodrug of choice be specifically activated in the tumor, but the source of the catalyst, be it bacterial, rodent, or human, will not be important. In fact, in the case of DT-diaphorase and mitomycin C, the rat form of the enzyme could be advantageous because it is more effective in activating mitomycin C than is the human form of this enzyme. Assuming targeted gene delivery to malignant cells, a non-host enzyme which is more effective at activating mitomycin C than the analogous host enzyme might also result in less drug activation in normal tissue and, hence, less normal tissue toxicity.


Assuntos
Mitomicinas/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Animais , Antibióticos Antineoplásicos/metabolismo , Biotransformação , Células CHO , Fracionamento Celular , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Transporte de Elétrons/fisiologia , Glutationa Transferase/metabolismo , Humanos , Porfiromicina/metabolismo , Ratos , Transfecção/genética
12.
Radiat Oncol Investig ; 5(5): 235-45, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9372546

RESUMO

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.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Porfiromicina/uso terapêutico , Antibióticos Antineoplásicos/efeitos adversos , Carcinoma de Células Escamosas/mortalidade , Terapia Combinada , Fracionamento da Dose de Radiação , Seguimentos , Neoplasias de Cabeça e Pescoço/mortalidade , Humanos , Mitomicina/efeitos adversos , Mitomicina/uso terapêutico , Porfiromicina/efeitos adversos , Taxa de Sobrevida , Fatores de Tempo
13.
Bioconjug Chem ; 7(6): 659-69, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8950485

RESUMO

Mitomycin C (1) is the prototypical bioreductive alkylating agent. Studies have shown that mitomycin C and its derivatives selectively alkylate guanine residues within di- and trinucleotide DNA sequences. This investigation sought to improve the selective DNA bonding properties of the mitomycins by coupling them with antisense oligodeoxynucleotides. Two procedures were developed that allowed the attachment of a phosphorothioate oligodeoxynucleotide containing a hexylamino spacer at the 5' terminus with a C(10)-activated mitomycin. In the first procedure, decarbamoylation of 1 (NaOCH3/ benzene) afforded 10-decarbamoylmitomycin C (10), which was treated with either dimethyl sulfate or methylthiochloroformate and base to yield 10-decarbamoylporfiromycin (11) and N(1a)-[(methylthio)-carbonyl]-10-decarbamoylmitomycin C (12), respectively. Activation of the C(10) site in 11 and 12 with 1,1'-carbonyldiimidazole or with 1,1'-thiocarbonyldiimidazole provided the N(1a)-substituted mitomycin 10-decarbamoyl-10-O-carbonylimidazoles (5, 7) and 10-decarbamoyl-10-O-thiocarbonylimidazoles (6, 8), respectively. Compounds 5-8 were reacted with glycine methyl ester hydrochloride (17) and base in both methylene chloride and aqueous buffered solutions to determine the ease and efficiency in which these C(10)-activated mitomycin derivatives coupled to amines. It was found that 5-8 all reacted with 17 in methylene chloride to give the coupled products 18-21 but that improved amine coupling yields in water were observed for the 10-decarbamoyl-10-O-thiocarbonylimidazoles 6 and 8 as compared with the 10-decarbamoyl-10-O-carbonylimidazoles 5 and 7. This finding led to the coupling of the phosphorothioate oligodeoxynucleotide, H2N(CH2)6-P(S)(OH)-GGCCCCGTG-GTGGCTCCAT (22) to 8. Compound 22 complemented a 19-base sequence in the translation initiation region of the human A-raf-1 gene. Use of excess 8 (28 equiv) with 22 gave only a 36% yield of the coupled product 23, which proved difficult to separate from 22. In the second procedure, phosphorothioate oligodexynucleotides that contained a hexylamino spacer at the 5'termini were coupled to 10-des(carbamoyloxy)-10-isothiocyanatoporfiromycin (9). Compound 9 was prepared in four steps from 11. Mesylation (methanesulfonyl chloride/pyridine) of 11 gave the C(10) mesylate 13, which was then treated with NaN3 (dimethylformamide, 90 degrees C) to give 10-des(carbamoyloxy)-10-azidoporfiromycin (14). Catalytic reduction (PtO2, H2) of 14 in pyridine afforded C(10) amine 15. Treatment of 15 with di-2-pyridyl thionocarbonate provided the desired 10-des(carbamoyloxy)-10-isothiocyanatoporfiromycin (9). Compound 9 readily coupled with 17 and base in both methylene chloride and aqueous buffered solutions to give 25. Use of the 5'hexylaminophosphorothioate oligodeoxynucleotides 32-35 in place of 17 gave the conjugated adducts 28-31, respectively, in a 12% to near-quantitative yield. The products were purified by semipreparative HPLC. Antisense agents 28-31 were designed to target a 30-base-long region from the coding region of the human FGFR1 gene. One adduct, 29, reduced the number of FGFR1 receptors in human aortic smooth cells for bFGF on the cell surface, which suggested down-regulation of FGFR1 gene expression. Further, 29 inhibited cultured human aortic smooth muscle cell proliferation and was less cytotoxic than porfiromycin (2). The biological assay data suggest that the phosphorothioate oligodexynucleotide porfiromycin conjugates may be more target selective and less toxic than either mitomycin or porfiromycin and thus be promising therapeutic agents.


Assuntos
Mitomicinas , Oligonucleotídeos/síntese química , Compostos Organotiofosforados/síntese química , Porfiromicina/análogos & derivados , Cromatografia Líquida de Alta Pressão , Humanos , Espectroscopia de Ressonância Magnética , Mitomicina/química , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Compostos Organotiofosforados/química , Porfiromicina/química , Porfiromicina/farmacologia
14.
Radiother Oncol ; 39(1): 65-71, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8735495

RESUMO

N-7[2-(4-nitrophenyldithio)-ethyl] mitomycin C, (BMS-181174; previously designated as BMY25067) is a mitomycin C analog now in initial clinical trials. The experiments described in this report were performed to assess whether BMS-181174, like mitomycin C and porfiromycin, was selectively toxic to the hypoxic cells in solid tumors and might therefore prove valuable in combination with radiotherapy. In contrast to mitomycin C and porfiromycin, BMS-181174 was more toxic to aerobic EMT6 cells in vitro than to cells made acutely hypoxic. In vitro, BMS-181174 and radiation produced cytotoxicity compatible with either additive or slightly supra-additive cytotoxicity. In vivo, BMS-181174 was effective in killing cells in solid EMT6 tumors. The effects of regimens combining BMS-181174 and radiation in vivo were complex. Combinations of low doses of BMS-181174 plus a large dose of radiation were very effective in killing cells in solid tumors. However, the survival curve plateaued at high doses of BMS-181174, providing evidence for a subpopulation of tumor cells which were resistant to both BMS-181174 and radiation; this was hypothesized to be a hypoxic cell population.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Mamárias Experimentais/radioterapia , Mitomicinas , Aerobiose/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos Alquilantes/administração & dosagem , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimioterapia Adjuvante , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Resistencia a Medicamentos Antineoplásicos , Feminino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Mitomicina/administração & dosagem , Mitomicina/uso terapêutico , Transplante de Neoplasias , Porfiromicina/uso terapêutico , Radiossensibilizantes/administração & dosagem , Radiossensibilizantes/uso terapêutico , Dosagem Radioterapêutica , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/radioterapia , Organismos Livres de Patógenos Específicos , Células Tumorais Cultivadas
15.
Proc Natl Acad Sci U S A ; 93(1): 456-60, 1996 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-8552660

RESUMO

Purified NADPH:cytochrome c (P-450) reductase (FpT; NADPH-ferrihemoprotein oxidoreductase, EC 1.6.2.4) can reductively activate mitomycin antibiotics through a one-electron reduction to species that alkylate DNA. To assess the involvement of FpT in the intracellular activation of the mitomycins, transfectants overexpressing a human FpT cDNA were established from a Chinese hamster ovary cell line deficient in dihydrofolate reductase (CHO-K1/dhfr-). The parental cell line was equisensitive to the cytotoxic action of mitomycin C under oxygenated and hypoxic conditions. In contrast, porfiromycin was considerably less cytotoxic to wild-type parental cells than was mitomycin C in air and markedly more cytotoxic under hypoxia. Two FpT-transfected clones were selected that expressed 19- and 27-fold more FpT activity than the parental line. Levels of other oxidoreductases implicated in the activation of the mitomycins were unchanged. Significant increases in sensitivity to mitomycin C and porfiromycin in the two FpT-transfected clones were seen under both oxygenated and hypoxic conditions, with the increases in toxicity being greater under hypoxia than in air. These findings demonstrate that FpT can bioreductively activate the mitomycins in living cells and implicate FpT in the differential aerobic/hypoxic toxicity of the mitomycins.


Assuntos
Mitomicina/toxicidade , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Porfiromicina/toxicidade , Aerobiose , Animais , Biotransformação , Células CHO , Cricetinae , Glutationa Transferase/metabolismo , Humanos , Hipóxia , Mitomicina/química , Mitomicina/metabolismo , Oxirredução , Porfiromicina/química , Porfiromicina/metabolismo , Proteínas Recombinantes
16.
Environ Mol Mutagen ; 25(1): 4-11, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7875125

RESUMO

The immediate effects of a single dose of the chemotherapeutic DNA crosslinking agent, mitomycin C (MMC), on the expression of several constitutive and drug-inducible genes were examined in a simple in vivo system, the 14 day chick embryo. We observed no effect of MMC on the steady-state mRNA expression of the constitutively expressed beta-actin, transferrin, or albumin genes. In contrast, MMC treatment significantly altered both the basal and drug-inducible mRNA expression of two glutethimide-inducible genes, 5-aminolevulinic acid (ALA) synthase and cytochrome P450 CYP2H1. The basal expression of these genes was transiently but significantly increased over a 24 hr period following a single dose of MMC. Conversely, MMC significantly suppressed the glutethimide-inducible expression of these genes when administered 1 to 24 hr prior to the inducing drug. The effects of MMC on both basal and drug-inducible ALA synthase and CYP2H1 mRNA expression were principally a result of changes in the transcription rates of these genes. In contrast, MMC treatment had little or no effect on glutethimide-induced expression of ALA synthase or CYP2H1 when administered 1 hr after the inducing drug, suggesting that a very early event in the induction process represents the target for these MMC effects. Covalent binding studies demonstrated that the effects of MMC on gene expression were closely correlated temporally with formation of [3H]-porfiromycin-DNA adducts. These results support the hypothesis that genotoxic chemicals specifically target their effects to inducible genes in vivo.


Assuntos
Reagentes de Ligações Cruzadas/farmacologia , Mitomicina/farmacologia , 5-Aminolevulinato Sintetase/genética , Animais , Embrião de Galinha , Sistema Enzimático do Citocromo P-450/genética , Indução Enzimática/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fígado/enzimologia , Porfiromicina/metabolismo , RNA Mensageiro/genética
17.
Adv Enzyme Regul ; 35: 117-30, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7572339

RESUMO

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.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Hipóxia Celular , Neoplasias Mamárias Experimentais/patologia , Porfiromicina/farmacologia , Animais , Antibióticos Antineoplásicos/farmacocinética , Sobrevivência Celular/efeitos dos fármacos , Radicais Livres/metabolismo , Radical Hidroxila/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mitomicina/farmacocinética , Mitomicina/farmacologia , Estrutura Molecular , NADPH-Ferri-Hemoproteína Redutase , Oxirredutases/metabolismo , Consumo de Oxigênio , Porfiromicina/farmacocinética
18.
Int J Radiat Oncol Biol Phys ; 29(2): 231-8, 1994 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8195012

RESUMO

The activity of three different classes of bioreductive drug, i.e., heterocyclic nitro compounds, N-oxides and quinones are compared. The major characteristics of RB-6145, tirapazamine and E09 are summarized and future directions for development of new bioreductive drugs are outlined. The concept of potentiating bioreductive drug activity by increasing tumor hypoxia is described and illustrated in particular by the use of photodynamic therapy (PDT) in combination with RSU-1069. Examples of how the therapeutic effectiveness of this approach can be studied by the use of 31P magnetic resonance spectroscopy is described. The effects of manipulation of nitric oxide (NO) levels in tumors by the use of modifiers of NO-synthase activity is illustrated by studies with the inhibitor nitro-L-arginine in experimental tumors. Associated changes in tumor physiology indicate promise for potential applications in therapy. Finally, changes in expression of reductase enzyme levels are considered in the context of the heterogenous nature of the tumor microenvironment.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Pró-Fármacos/uso terapêutico , Animais , Humanos , Óxido Nítrico/metabolismo , Nitroimidazóis/uso terapêutico , Oxirredução , Fotoquimioterapia , Porfiromicina/uso terapêutico , Tirapazamina , Triazinas/uso terapêutico
19.
Cell Prolif ; 27(3): 153-63, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10465006

RESUMO

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.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias Mamárias Experimentais/patologia , Mitomicina/farmacologia , Porfiromicina/farmacologia , Aerobiose , Anaerobiose , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Feminino , Camundongos , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
20.
Mol Pharmacol ; 44(6): 1128-34, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8264549

RESUMO

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.


Assuntos
Mitomicina/farmacologia , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , Porfiromicina/farmacologia , Animais , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel de Poliacrilamida , Humanos , Concentração de Íons de Hidrogênio , Cinética , NAD/metabolismo , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...