Please use this identifier to cite or link to this item: https://rima.ufrrj.br/jspui/handle/20.500.14407/10258
Full metadata record
DC FieldValueLanguage
dc.contributor.authorBernardes, Bauer de Oliveira
dc.date.accessioned2023-12-21T18:59:41Z-
dc.date.available2023-12-21T18:59:41Z-
dc.date.issued2012-11-21
dc.identifier.citationBERNARDES, Bauer de Oliveira. Síntese de derivados das quinonas lapachol, α- e β-lapachona. Explorando o sistema quinônico. 2012. 348 f. Tese (Doutorado em Química) - Instituto de Ciências Exatas, Universidade Federal Rural do Rio de Janeiro, Seropédica, 2012.por
dc.identifier.urihttps://rima.ufrrj.br/jspui/handle/20.500.14407/10258-
dc.description.abstractNeste trabalho exploramos a síntese de dioxolanos, dioxinas, ésteres, éteres e fenazinas a partir das quinonas lapachol, α-lapachona, 2-metóxi-lapachol e, principalmente, β-lapachona. Verificamos que a β-lapachona pode formar dioxolano a partir de diazompostos ou nitroalcanos. Estudamos reações fotoquímicas entre o-quinonas e diferentes sistemas olefínicos, onde observamos cicloadições do tipo [4+2], formação de um aldeído inédito na reação com 2,3-di-hidro-1,4-dioxina, e a característica da β-lapachona em transferir energia através da dimerização do acenaftileno. Além disso, fotólise por pulso de laser das quinonas α-, β-, nor-β- e 3-sulfo-β-lapachona foi usada para caracterizar e estudar a reatividade e propriedades do estado excitado triplete destas quinonas. Reações de redução seguidas de acetilação ou alquilação foram realizadas com lapachol, α-, β-lapachona e 2-metóxi-lapachol, e o uso de piridina nesta última quinona forneceu principalmente a α-xiloidona. A β-lapachona submetida ao NaBH4 ou LiAlH4 seguido de brometo de benzila forneceu um álcool terciário, só obtido anteriormente por nosso grupo, através de reação de Grignard. Novas fenazinas e quinoxalinas foram formadas por reações com diaminas vicinais. Aplicamos metodologias distintas na tentativa de nitração dos ésteres, éteres, dioxina ou dioxolano formados a partir das quinonas, mas em quase todos os casos houve retorno a forma quinônica; Nas condições de Laszlo uma dioxina formou um acetal inédito. Os compostos obtidos foram analisados por espectrometria de massa, espectroscopia de ressonância magnética nuclear de carbono-13 e hidrogênio-1, infravermelho e/ou difração de raio-X.por
dc.description.sponsorshipConselho Nacional de Desenvolvimento Científico e Tecnológico, CNPq, Brasil.por
dc.formatapplication/pdf*
dc.languageporpor
dc.publisherUniversidade Federal Rural do Rio de Janeiropor
dc.rightsAcesso Abertopor
dc.subjectβ-lapachonapor
dc.subjectnaftoquinonaspor
dc.subjectcicloadiçõespor
dc.subjectβ-lapachoneeng
dc.subjectnaphthoquinoneseng
dc.subjectcycloadditioneng
dc.titleSíntese de derivados das quinonas lapachol, α- e β-lapachona. Explorando o sistema quinônicopor
dc.title.alternativeSynthesis of quinone derivatives of lapachol, α- and β-lapachone. Exploring the system quinoneeng
dc.typeTesepor
dc.description.abstractOtherIn this work we investigated the reactivity of the quinones lapachol, -lapachone, 2- methoxy-lapachol and mainly -lapachone. It was found that diazocompounds react with -lapachone with formation of dioxolanes, but with diazomethane an oxirane and an aldehyde were also formed, depending on the reaction conditions. Dioxolanes were also made from nitroalkanes by reaction with -lapachone, but nitromethane was unreactive. We have made theoretical calculations to compare the reactivity of different nitroalkanes and Grignard compounds with this quinone. Reactions with vicinal diamines led to different phenazines and quinoxalines, some unpublished. Photochemical reactions with different systems olefinic were observed in type cycloaddition [4 +2], notably the reaction with 5,6-diphenyl-2,3-dihydro-1,4-dioxin product which provided a yield of 96% and we not isolate products by cycloaddition [2 +2]. We observed the characteristic of -lapachone in transferring energy through the dimerization of acenaphthylene and also isolated a novel aldehyde in the reaction with 2,3-dihydro-1 ,4-dioxin. Laser flash photolysis of quinones -, -, nor-- and 3-sulfo-- lapachone was used to characterize and study the properties and reactivity of the triplet excited state of these quinones. Different vicinal diamines reacted with -lapachone to form new quinoxalines and phenazines. Reduction reactions followed by acetylation or alkylation were performed with lapachol, -, -lapachone and 2-methoxy-lapachol, and the use of pyridine latter quinone provided mainly -xiloidona. The -lapachone subjected to LiAlH4 or NaBH4 followed by benzoyl bromide gave a tertiary alcohol, previously only obtained via Grignard reaction by our group. Distinct methodologies were used in attempting the nitration products obtained, such as esters, ethers or dioxin or dioxolane ring, but in almost all cases there was a return to quinone form. Under the conditions of Laszlo dioxin DXBL formed a novel acetal. The compounds obtained were analyzed by mass spectrometry, nuclear magnetic resonance spectroscopy of carbon-13 and hydrogen-1, infrared and / or X-ray diffraction.eng
dc.contributor.advisor1Ferreira, Aurélio Baird Buarque
dc.contributor.advisor1ID237.924.007-82por
dc.contributor.advisor1Latteshttp://lattes.cnpq.br/5526484175547597por
dc.contributor.referee1Silva, Francisco de Assis da
dc.contributor.referee2Mello, Heloísa de
dc.contributor.referee3Ferreira, José Carlos Netto
dc.contributor.referee4Garden, Simon John
dc.creator.ID041.532.457-64por
dc.creator.Latteshttp://lattes.cnpq.br/8988332784422026por
dc.publisher.countryBrasilpor
dc.publisher.departmentInstituto de Ciências Exataspor
dc.publisher.initialsUFRRJpor
dc.publisher.programPrograma de Pós-Graduação em Químicapor
dc.relation.referencesABREU, F.C., FERREIRA, D.C.M., GOULART, M.O.F., BURIEZ, O. e AMATORE, C.. Eletrochemical activation of -lapachone in -cyclodextrin inclusion complexes and reactivity of its reduced form towards oxygen in aqueous solutions – J. Electroanal. Chem., 608, p125-132, 2007. ABREU, F.C., FERREIRA, D.C.M., WADHAWAN, J., AMATORE, C., FERREIRA, V.F., SILVA, M.N., SOUZA, M.C.B.V., GOMES, T.S., XIMENES, E.A. e GOULART, M.O.F.. Eletrochemistry of -lapachone and its diazoderivative: Relevance to their compared antimicrobial activites – Electrochem. Commun., 7, p767-72, 2005. AL-AZMI, A., ELASSAR, A.Z.A. e BOOTH, B. L.. The chemistry of diaminomaleonitrile and its utility in heterocyclic synthesis. Tetrahedron, 59, p2749-63, 2003. ALEGRÍA, A.E., SANCHEZ-CRUZ, P. e RIVAS, L.. Alkaline-earth cátions enhance ortho-quinone-catalyzed ascorbate oxidation. Free Radical Biol. Med., 37, p1631-9, 2004. ALI, R. M. , HOUGHTON, P. J. e HOO, T. S.. Antifungal activity of some bignoniacea found in Malaysia. Phytother. Res., 12(5), p331-4, 1998. ALMEIDA, L.S., VILLEGAS, R.A.S. e RIBEIRO, C.M.. Estudo da ciclofuncionalização do lapachol com triacetato de tálio. In: 24ª RA-SBQ, Poços de Caldas, 2001. ALVES, G.B.C., LOPES, R.S.C., LOPES, C.C. E SNIECKUS, V. – Two expedient syntheses of -lapachone, Synthesis-Stuttgart, 11, p1875-7, 1999. ALVES, G.B., ROLIM, L.A., ROLIM NETO, P.J., LEITE, A.C.L., BRONDANI, D.J., DE MEDEIROS, F.P.M., BIEBER, L.W. e MENDONÇA JÚNIOR, F.J.B. – Purificação e caracterização da -lapachona e estudo de estabilidade dos cristais em diferentes condições de armazenamento, Quim. Nova, 31 (2), p413-6, 2008. AMARAL, A.C.F. e BARNES, R.A.. The total synthesis of -lapachone. J. Heterocycl. Chem., 29 (6), p1457-60, 1992. ANDRADE-NETO, V.F., GOULART, M.O.F., FILHO, J.F.S., SILVA, M.J., PINTO, M.C.F.R., PINTO, A.V.,ZALIS, M.G., CARVALHO, L.H. e KRETTLIA, A.U.. Antimalarial activity of phenazines from lapachol, -lapachone and its derivatives against Plasmodium falciparum in vitro and Plasmodium berghei “in vivo”. Bioorg. Med. Chem. Lett., 14, p1145–9, 2004. ANTKOWIAK, W.Z. e SOBCZAK, A.. Solvent effect on the reactivity of 1,10- phenanthroline-5,6-dione towards diazomethane. Tetrahedron, 57, p2799-805, 2001. ANTUNES, R.M.P., LIMA, E.O., PEREIRA, M.S.V., CÂMARA, C.A., ARRUDA, T.A., CATÃO, R.M.R., BARBOSA, T.P., NUNES, X.P., DIAS, C.S. e SILVA, T.M.S.. Atividade antimicrobiana “in vitro” e determinação da concentração inibitória mínima (CIM) de fitoconstituintes e produtos sintéticos sobre bactérias e fungos leveduriformes. Rev. Bras. Farmacogn., 16(4), p517-23, 2006. ARAÚJO, E.V., ALENCAR, J.R.B. e NETO, P.J.R.. Lapachol: segurança e eficácia na terapêutica. Rev. Bras. Farmacogn., 12 (supl.), p57-9, 2002. BALASSIANO, I.T., DE PAULO, S.A., SILVA, N.H., CABRAL, M.C. e CARVALHO, M.G.C.. Demonstration of the lapachol as a potencial drug for reducing cancer metastasis. Oncol. Rep., 13, p329-33, 2005. 154 BARBOSA, T.C., CAMARA, C.A., SILVA, T.M.S., MARTINS, R.M., PINTO, A.C. e VARGAS, M.D.. New 1,2,3,4-tetrahydro-1-aza-anthraquinones and 2-aminoalkyl compounds from norlapachol with molluscicidal activity. Bioorg. Med. Chem., 13, p6464-9, 2005. BARREIRO, E.J. e FRAGA, C.A.M.. Química Medicinal – As bases moleculares da ação dos fármacos. 1ª ed., Ed. Artmed, São Paulo, 2001. BECKER, H. G. O., BERGER, W., DOMSCHKE, G., FANGHÄNEL, E. e FAUST, J., ORGANIKUM – Química Orgânica Experimental. 2ª ed., Lisboa, Fundação Calouste Gulbenkian, 1997. BEDIR, E., PEREIRA, A.M.S., KHAN, S.I., CHITTIBOYINA, A., MORAES, R.M. e KHAN, I.A. – “A new -lapachone derivative from Distictella elongata (Vahl) Urb., J. Braz. Chem. Soc., 20 (2), p383-6, 2009. BENTLE, M.S., BEY, E.A., DONG, Y., REINICKE, K.E. e BOOTHMAN, D.A.. New tricks for old drugs: the anti carcinogenic potencial of DNA repair inhibitors, J. Mol. Histol., 37, p203-18, 2006a. BENTLE, M.S., REINICKE, K.E., BEY, E.A., SPITZ, D.R. e BOOTHMAN, D.A.. Calcium-dependent modulation of poly(ADP-ribose) polymerase-1 alters cellular metabolism and DNA repair, J. Biol. Chem., 281 (44), p33684-96, 2006b. BEKELE, T., SHAH, M.H., WOLFER, J., ABRAHAM, C.J., WEATHERWAX, A. e LECTKA, T.- Catalytic, enantioselective [4 + 2]-cycloadditions of ketene enolates and o-quinones: Efficient entry to chiral, -oxygenated carboxylic acid derivatives, J. Am. Chem. Soc., 128, p1810, 2006. BERNARDES, BAUER DE OLIVEIRA. Alguns derivados da -lapachona. Dissertação (Mestrado em Ciências) Programa de Pós-Graduação em Química Orgânica. 2001. 91f. Universidade Federal Rural do Rio de Janeiro, Seropédica, 2001. BERNARDES, B.O., FERREIRA, A.B.B. e SILVA, A.R.. Estudos sobre -lapachona e alguns derivados. In: 23ª RA-SBQ, 2000, Poços de Caldas. BERNARDES, B.O., FERREIRA, A.B.B.. Novas fenazinas obtidas de -lapachona. In: 27ª RA-SBQ, Salvador, 2004. BERNARDES, B.O., SILVA, A.R., FERREIRA, A.B.B. e SANT'ANNA, C. M. R. . Estudo de reações de adição por nitroalcanos e reagentes de Grignard à onaftoquinona beta-lapachona, utilizando método semi-empírico PM3. In: 29a Reunião anual da Socidade Brasileira de Química, 2006, Águas de Lindóia-SP. BEY, E.A., BENTLE, M.S., REINICKE, K.E., DONG, Y, YANG, C-R., GIRARD, L., MINNA, J.D., BORNMANN, W.G., GAO, J. e BOOTHMAN, D.A.. An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by -lapachone, Proc. Natl. Acad. Sci. U.S.A., 104 (28), p11832-7, 2007. BEY, E.A., WUERZBERGER-DAVIS, S.W., PINK, J.J., YANG, C.R., ARAKI, S., REINICKE, K.E., BENTLE, M.S., DONG, Y., CATALDO, E., CRISWELL, T.L., WAGNER, M.W., LI, L., GAO, J. e BOOTHMAN, D.A.. Morning with art, lessons learned: Feedback regulation, restriction threshold biology, and redundancy govern molecular stress responses, J. Cell. Physiol., 209, p604-10, 2006. BINUTU, O.A., ADESOGAN, K.E. e OKOGUN, J.I.. Antibacterial and antifungal compounds from Kigelia pinnata, Planta Med., 62, p352-3, 1996. BLANCO, E., BEY, E.A., DONG, Y., WEINBERG, B.D., SUTTON, D.M., BOOTHMAN, D.A. e GAO, J.- -lapachone-containing PEG-PLA polymer micelles as novel nanotherapeutics against NQO1-overexpressing tumor cells. J. Controlled Release, 122, p365-74, 2007. 155 BONIFAZI, E.F., RÍOS-LUCI, C., LEÓN, L.G., BURTON, G., PADRÓN, J.M., MISICO, R.I. - Antiproliferative activity of synthetic naphthoquinones related to lapachol. First synthesis of 5-hydroxylapachol, Bioorg. Med. Chem., 18, p2621-30, 2010. BOORSTEIN, R.J. e PARDEE, A.B.. -lapachone greatly enhances mms lethality to human fibroblasts, Biochem. Biophys. Res. Commun., 118, p828-34, 1984. BOOTHMAN, D.A.. Enhanced malignant transformation is accompanied by increased survival recovery after ionizing radiation in Chinese hamster embryo fibroblasts, Radiat. Res., 138, S121-5, 1994. BOOTHMAN, D.A., FUKUNAGA, N. E WANG, M.. Down-regulation of topoisomerase I in mammalian cells following ionizing radiation, Cancer Res., 54, p4618-26, 1994. BOOTHMAN, D.A., GREER, S. E PARDEE, A.B.. Potentiation of halogenated pyrimidine radiosensitizers in human carcinoma cells by -lapachone (3,4-dihydro- 2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione), a novel DNA repair inhibitor, Cancer Res., 47, p5361-6, 1987. BOOTHMAN, D.A. e PARDEE, A.B.. Inhibition of radiation-induced neoplastic transformation by -lapachone, Proc.Natl. Acad. Sci. U.S.A., 86, p4963-7, 1989. BOOTHMAN, D.A., TRASK, D.K. E PARDEE, A.B.- Inhibition of potentially lethal DNA damage repair in human tumor cels by -lapachone, an activator of topoisomerase I, Cancer Res., 49, p605-12, 1989. BOVERIS, A., DOCAMPO, R., TURRENS, J.F. e STOPPANI, O.M.. Effect of - lapachone on superoxide anion and hydrogen peroxide production in Trypanosoma cruzi, Biochem. J., 175(2), p431-9, 1978. BRANDÃO, M.A.F., DE OLIVEIRA, A.B. e SNIECKUS, V.. Combined directed metalation. cross coupling strategies. A regiospecific route to heteroring-annelated ortho-naphthoquinones and a short synthesis of -lapachona - Tetrahedron Lett., 34(15), p2437-40, 1993. BRUNETON, J.. Pharmacognosy, phytocehmistry and medicinal plants, Ed. Tecnique & Documentation. Lavoisier, 1995. CAMARA, C.A., PINTO, A.C., ROSA, M.A. e VARGAS, M.D.. Secondary amines and unexpected 1-aza-anthraquinones from 2-methoxylapachol, Tetrahedron, 57, p9569-74, 2001. CAMARA, C.A., PINTO, A.C., VARGAS, M.D. e ZUKERMAN-SCHCEPTOR, J.. Azepines from intramolecular Prins cyclization of an aminodrivative of lapachol, Tetrahedron, 58, p6135-40, 2002. CAMPOS-TAKAKI, G.M., STEIMAN, R., SEIGILE-MURANDI, F., SILVA, A.A. e BIEBER, L.. Effect of médium composition on the antifungal susceptibility tests to lapachol, -lapachone, phthiocol and its synthetic analogues, Rev. Microbiol., 23(2), p106-11, 1992. CASTELLANOS, J.R.G, PRIETO, J.M. E HEINRICH, M. - Red Lapacho (Tabebuia impetiginosa) - A global ethnopharmacological commodity?, J. Ethnopharmacol., 121, p1-13, 2009. CARDOSO, S.R.F., de MOURA, K.C.G., EMERY, F.S., SANTANA, L.A., PINTO, C.N., PINTO, M.C.F.R. e PINTO, A.V.. Synthesis of some naturally occurring naphthoquinones from Paracetoma and Tabebuia species - An. Acad. Bras. Ciênc., 69 (1), p15-8, 1997. CARNEIRO, P.F., PINTO, M.C.F.R., COELHO, T.S., CAVALCANTI, B.C., PESSOA, C., DE SIMONE, C.A., NUNES, I.K.C., DE OLIVEIRA, N.M., DE ALMEIDA, R.G., PINTO, A.V., DE MOURA, K.C.G., DA SILVA, P.A. E DA 156 SILVA JÚNIOR, E.N. - Quinonoid and phenazine compounds: Synthesis and evaluation against H37Rv, rifampicin and isoniazid-resistance strains of Mycobacterium tuberculosis, Eur. J. Med. Chem., 46, p4521-9, 2001. CARVALHO, C.E.M., LUCAS, N.C., HERRERA, J.O.M., PINTO, A.V., PINTO, M.C.F.R., BRINN, I.M.. Fluorescent symmetric phhenazines from naphthoquinones 4. Solvent effect on time-resolved fluorescence J. Photochem. Photobiol., A, 167, p1-9, 2004. CHAU, Y.P., SHIAH, S.G., DON, M.J. e KUO, M.L.. Involvement of hydrogen peroxide in topoisomerase inhibitor -Lapachone-induced apoptosis and differentiation in human leukemia cells - Free Radical Biol. Med., 24 (4), p660-70, 1998. CHAVÉS, J.P., PINTO, M.C.F.R. e PINTO, A.V.. Heterocyclics from quinones, I – The reaction with primary alkyl amines. J. Braz. Chem. Soc., 1 (1), p22-7, 1990. CHOI, E.K, TERAI, K., JI, I.M., KOOK, Y.H., PARK, K.H., OH, E.T., GRIFFIN, R.J., LIM, B.U., KIM, J.S., LEE, D.S., BOOTHMAN, D.A., LOREN, M., SONG, C.W. e PARK, H.J.. Upregulation of NAD(P)H:Quinone Oxidoreductase By Radiation Potentiates the Effect of Bioreductive -Lapachone on Câncer Cells. Neoplasia, 9 (8), p634-42, 2007. CI, X., SILVA, R.S., GOODMAN, J.L., NICODEM, D.E. e WHITTEN, D.G.. A reversible photoredox reaction: Electron-transfer photoreduction of -Lapachone by triethylamine, J. Am. Chem. Soc., 110, p8548-50, 1988. CI, X., SILVA, R.S., NICODEM, D., WHITTEN, D.G.. Electron and hydrogen atom transfer mechanism for the photoreduction of o-quinones. Visible light induced photoreactions of -lapachone with amines, alcohols, and amino alcohols, J. Am. Chem. Soc., 111 (4), p1337-43, 1989. CI, X. e WHITTEN, D.G.. Photofragmentation via Single-Electron Transfer: Selective Labilization of C-C Bonds in Amino Alcohols with Several Bonds between Heteroatom Substituents - J. Am. Chem. Soc., 111, p3459-61, 1989. COCQUET, G., ROOL, P e FERROUD, C.- A catalytic versus stoichiometric photoinduced electron transfer promoted selective C16_C21 bond cleavage of catharanthine - Tetrahedron Lett., 42, p839–841, 2001. COELHO T.S., SILVA, R.S.F., PINTO, A.V., PINTO, M.C.F.R., SCAINI, C.J., MOURA, K.C.G e SILVA, P.A. - Activity of -lapachone derivatives against rifampicin-susceptible and –resistant strains of Mycobacterium tuberculosis. Tuberculosis, 90, p293-7, 2010. CORNÉLIS, A. e LASZLO, P., - Clay-Supported Copper(II) and Iron(III) Nitrates: Novel Multi-Purpose Reagents for Organic Synthesis, Synthesis, 10, p909-18, 1985. CORNÉLIS, A., DELAUDE, L., GERSTMANS, A. e LASZLO, P. - A procedure for quantitative regioselective nitration of aromatic hydrocarbons in the laboratory Tetrahedron Lett., 29, p5657-60, 1988. CÔRREA, G., VILELA, R., MENNA-BARRETO, R.F.S, MIDLEJ, V. e BENCHIMOL, M. – Cell death induction in Giardia lamblia: Effect of betalapachone and starvation, Parasitol. Int., 58, p424-37, 2009. COSTA, P.R.R. - safrol e eugenol: Estudo da reatividade química e uso em síntese de produtos naturais biologicamente ativos e seus derivados, Quím. Nova, 23 (3), p357- 69, 2000. CROMBIE, L., ROSSITER, J. T., BRUGGEN, V. e WHITING, D. A.. Deguelin cyclase a prenyl to chrome transforming enzyme from Tephrosia vogelli Phytochemistry, 31 (2), p451-61, 1992. 157 CRUZ, F.S., DOCMAPO, R. e BOVERIS, A.. Generation of superoxides anions and hydrogen peroxide from -lapachone in bacteria, Antimicrob. Agents Chemother., 14(4), p630-3, 1978. CRUZ, F.S., GILBERT, B., LOPES, J.N., PICHIN, R. e PINTO, A.V.. The reaction of - and -lapachone with N-bromosuccinimide - Rev. Latin. Quím., 8, p138-40, 1977. CUNHA, A.S.. Dissertação de mestrado. Espermidinas acopladas com 1,4-naftoquinonas. uma nova classe de substâncias com possível atividade anti-Tumoral. UFRJ, 2005. CUNHA-FILHO, M.S.S., DACUNHA-MARINHO, B., TORRES-LABANDEIRA, J.J., MARTÍNEZ-PACHECO, R. e LANDÍN, M.. Characterization of -lapachone and methylated -cyclodextrin solid-state systems. AAPS PharmSciTech, 8 (3), artigo 60, 2007. Acessado por http://www.aapspharmscitech.org/articles/pt0803/pt0803060/ pt0803060.pdf em 16/11/2010. CUNHA-FILHO, M.S.S., ÉSTEVEZ-BRAUN, A., PÈREZ-SACAU, E., ECHEZARRETA-LÓPEZ, M.M., MARTINEZ-PACHECO, R. e LANDIN, M.. Light effect on the stability of β-lapachone in solution: pathways and kinetics of degradation – J. Pharmacy and Pharmacology, 63, p1156-60, 2011. CUNHA-FILHO, M.S.S., LANDIN, M., MARTINEZ-PACHECO, R. e DACUNHAMARINHO, B.. -lapachone - Acta Crystallogr., Sect. C: Cryst. Struct. Commun., C62, p.o473-5, 2006. CUNHA-FILHO, M.S.S., MARTINEZ-PACHECO, R. e LANDÍN, M.. Compatibility of the antitumoral -lapachone with different solid dosage forms excipients. Journal of Pharmaceutical Analysis, 45, p590-8, 2007. CUNHA-FILHO, M.S.S., MARTINEZ-PACHECO, R. e LANDÍN, M.. – Dissolution rate enhacement of the novel antitumora -lapachone by solvent change precipitation of microparticles, Eur. J. Pharm. Biopharm., 69, p871-7, 2008. DA SILVA, F.C., FERREIRA, S.B., KAISER, C.R., PINTO, A.C. e FERREIRA, V.F. – Synthesis of α- and β-Lapachone Derivatives from Hetero Diels-Alder Trapping of Alkyl and Aryl o-Quinone Methides, J. Braz. Chem. Soc., 20 (8), p1478-82, 2009. DA SILVA, M.J., PINTO, MC.F.R., DE SIMONE, C.A., SOARES, J.G., REYS, J.R.M., DE SOUZA FILHO, J.D., HARRISON, W.T.A., CARVALHO, C.E.M., GOULART, M.O.F., DA SILVA JÚNIOR e PINTO, A.V. - Synthesis and characterisation of a new polycyclic phenazine from1,4-naphthoquinone, Tetrahedron Lett., 52, p2415-8, 2011. DA SILVA JÚNIOR, E.N., CAVALCANTI, B.C., GUIMARÃES, T.T., PINTO, M.C.F.R., CABRAL, I.O., PESSOA, C., COSTA-LOTUFO, L.V., DE MORAES, M.O., DE ANDRADE, C.K.Z., DOS SANTOS, M.R., DE SIMONE, C.A., GOULART, M.O.F. E PINTO, A.V. - Synthesis and evaluation of quinonoid compounds against tumor cell lines – Eur. J. Med. Chem., 46, p399-410, 2011. DA SILVA JÚNIOR, E.N., DE DEUS, C., CAVALCANTI, B.C., PESSOA, C., COSTA-LOTUFO, L.V., MONTENEGRO, R.C., DE MORAES, M.O., PINTO, M.C.F.R., SIMONE, C.A., FERREIRA, V.F., GOULART, M.O.F., ANDRADE, C.K.Z., e PINTO, A.V., 3-Arylamino and 3-Alkoxy-nor-β-lapachone Derivatives: Synthesis and Cytotoxicity against Cancer Cell Lines - J. Med. Chem., 53, p504– 508, 2010b. DA SILVA JÚNIOR, E.N., DE MOURA, M.A.B.F., PINTO, A.V., PINTO, M.C.F.R., DE SOUZA, M.C.B.V., ARAÚJO, A.J., PESSOA, C., COSTA-LOTUFO, L.V., MONTENEGRO, R.C., DE MORAES, M.O., FERREIRA, V.F. e GOULART, M.O.F., Cytotoxic, Trypanocidal Activities and Physicochemical Parameters of nor- β-Lapachone-based 1,2,3-Triazoles, J. Braz. Chem. Soc., 20 (4), p635-43, 2009b. 158 DA SILVA JÚNIOR, E.N., DE SIMONE, C.A., DE SOUZA, A.C.B., PINTO, C.N., GUIMARÃES, T.T., PINTO, M.C.F.R.,e PINTO, A.V. – Unexpected transformation of quinones to spirolactones and to naturally occurring naphthalenic compounds, Tetrahedron Lett., 50, p1550-3, 2009c. DA SILVA JÚNIOR, E.N., DE SOUZA, M.C.B.V., FERNANDES, M.C., MENNABARRETO, R.F.S., PINTO, M.C.F.R., LOPES, F.A., SIMONE, C.A., ANDRADE, C.K.Z., PINTO, A.V., SILVA, R.S.F., FERREIRA, V.F. e DE CASTRO, S.L., Synthesis and anti-Trypanosoma cruzi activity of derivatives from nor-lapachones and lapachones – Bioorg. Med. Chem., 16, p5030–8, 2008b. DA SILVA JÚNIOR, E.N., DE SOUZA, M.C.B.V., PINTO, A.V., PINTO, M.C.F.R, GOULART, M.O.F., BARROS, F.W.A., PESSOA, C., COSTA-LOTUFO, L.V. MONTENEGRO, R.C., DE MORAES, M.O. e FERREIRA, V.F., Synthesis and potent antitumor activity of new arylamino derivatives of nor-b-lapachone and nor-alapachone – Bioorg. Med. Chem., 15, p7035-41, 2007. DA SILVA JÚNIOR, E.N., GUIMARÃES, T.T., MENNA-BARRETO, R.F.S., PINTO, M.C.F.R., SIMONE, C.A., PESSOA, C., CAVALCANTI, B.C., SABINO, J.R., ANDRADE, C.K.Z., GOULART, M.O.F., CASTRO, S.L. e PINTO, A.V., The evaluation of quinonoid compounds against Trypanosoma cruzi: Synthesis of imidazolic anthraquinones, nor--lapachone derivatives and -lapachone-based 1,2,3-triazoles Bioorg. Med. Chem., 18, p3224-30, 2010a. DA SILVA JÚNIOR, E.N., MENNA-BARRETO, R.F.S., PINTO, M.C.F.R., SILVA, R.S.F., TEIXEIRA, D.V., DE SOUZA, M.C.B.V., DE SIMONE, C.A., DE CASTRO, S.L., FERREIRA, V.F. e PINTO, A.V., Naphthoquinoidal [1,2,3]- triazole, a new structural moiety active against Trypanosoma cruzi – Eur. J. Med. Chem., p1774-80, 2008a. DA SILVA JÚNIOR, E.N., PINTO, M.C.F.R., MOURA, K.C.G., SIMONE, C.A., NASCIMENTO, C.J., ANDRADE, C.K.Z. E PINTO, A.V., Hooker’s ‘lapachol peroxide’ revisited - Tetrahedron Lett., 50, p1575–7, 2009a. DAWSON, B.A., GIRARD, M., KINDACK, FILLION, J. e AWANG, D.V.C.. 13C NMR of lapachol and some Related naphthoquinones - Magn. Reson. Chem., 27 (12), p1176-83, 1989; DE OLIVEIRA, A. B., RASLAN, D. S., DE OLIVEIRA, G. e MAIA, J. G. S.. Lignans and naphthoquinones from Tabebuia incana, Phytochemistry, 34 (5), p1409-12, 1993. DE OLIVEIRA, A. B., RASLAN, D. S., KHUONG-HUU, F.. Selenium reagent in the synthesis of naphtho[2,3-b]furan-4,9-diones - Tetrahedron Lett., 31 (47), p6873-6, 1990a. DE OLIVEIRA, A. B., RASLAN, D. S., MIRAGLIA, C. M., MESQUITA, A. A. L., ZANI, C. L., FERREIRA, C. T. e MAIA, J. G.S.. Estrutura química e atividade biológica de naftoquinonas de bignoniáceas brasileiras - Quím. Nova 13 (4) p302-7, 1990b. DEGRASSI, F., SALVIA, R. e BERGHELLA, L.. The production of chromosomal alterations by -lapachone an activator of topoisomerase I, Mutat. Res., 288, p263- 7, 1993. DETERDING, A., DUNGEY, F.A., THOMPSON, K.A. e STEVERDING, D.. Antitrypanosomal activities of DNA topoisomerase inhibitors, Acta Trop., 93, p311-6, 2005. DI CHENNA, P.H., BENEDETTI-DOCTOROVICH, V., BAGGIO, R.F., GARLAND, M.T. E BURTON, G.. Preparation and citotoxicity toward cancer cells mono(arylimino) derivatives of -lapachone, J. Med. Chem., 44, p2486-9, 2002. 159 DOCAMPO, R., CRUZ, F.S., BOVERIS, A., MUNIZ, R.P.A. e ESQUIVEL, D.M.S.. Lipid peroxidation and the generation of free radicals, superoxide anion, and hydrogen peroxide in -lapachone-treated Trypanosoma cruzi epimastigotes. Arch. Biochem. Biophys., 186(2), p292-7, 1978. DOCAMPO, R., CRUZ, F.S., BOVERIS, A., MUNIZ, R.P.A. e ESQUIVEL, D.M.S.. -lapachone enhacement of lipid peroxidation and superoxide anion and hydrogen peroxide formation by sarcoma 180 ascites tumor cells - Biochem. Pharmacol., 28 (6), p723-8, 1979. DOCAMPO, R., LOPES, J.N., CRUZ, F.S. e SOUZA, W.. Tripanosoma cruzi: Ultrastructural and metabolic alterations of epimastigotes by -lapachone – Exp. Parasitol, 42, p142-9, 1977. DOCTOROVICH, V.B-, ESCOLA, N. e BURTON, G.. Preparation and NMR characterization of new substituited benzo[a]phenazines. Magn. Reson. Chem., 36 (7), p529, 1998. DON, M.J., CHANG, Y.H., CHEN, K.K., HO, L.K. e CHAU, Y.P.. Induction of CDK Inhibitors (p21WAF1 and p27Kip1) and Bak in the -Lapachone-Induced Apoptosis of Human Prostate Cancer Cells. Mol. Pharmacol., 59 (4), p784-94, 2001. DONNELLY, D M. e O’REILLY, J. 6-methyldehydro--lapachone from Fomes annosus, Phytochemistry, 19 (2), p277-9, 1980. DUBIN, M., VILLAMIL, S.H.F. e STOPPANI. Citotoxicidad de la -lapachona: una onaftoquinona con posibles usos terapéuticos, Medicina-Buenos Aire, 61, p343-50, 2001. DUDLEY, K.H. e MILLER, H.W.. The mercuric acetate oxidation of isolapachol. J. Am. Chem. Soc., 32, p241-4, 1967. EISNER, T. e ANESHANSLEY, D.J.. Spray aiming in the bombardier beetle: Photographic evidence - Proc. Natl. Acad. Sci. U.S.A., 96, p9705–9, 1999. ESTEVES-SOUZA, A., FIGUEIREDO, D.V., ESTEVES, CAMARA, C.A., VARGAS, M.D., PINTO, A.C., e ECHEVARRIA, A.. Cytotoxic and DNA-topoisomerase effects of lapachol amine derivativesand interactions with DNA - Braz. J. Med. Biol. Res, 40(10), p1399-1402, 2007. EYONG, K.O., KUMAR, P.S., KUETE, V., FOLEFOC, G.N., NKENGFACK, E.A. e BASKARAN, S. – Semisynthesis and antituomral activity of 2- acetylfuranonaphthoquinone and other naphthoquinone derivatives from lapachol, Bioorg. Med. Chem. Lett., 18, p5387-90, 2008. FALKENBERG, M. B.. Farmacognosia da planta ao medicamento, p545-570 Organizadores: SIMÕES, C. M. O., MELLO, J.C.P., SCHENKEL, E.P., GOSMANN, G., MENTZ, L.A. e PETROVICK, P.R., Ed. Universidade Federal de Santa Catarina e Universidade Federal do Rio Grande do Sul. 1ª ed., 1999. FELÍCIO, A.C., CHANG, C.V., BRANDÃO, M.A., PETERS, V.M. e GUERRA, M.O.. Fetal growth in rats treated with lapachol - Contraception, 66, p289-93, 2002. FERRAZ, P.A.L., ABREU, F.C., PINTO, A.V., GLEZER, V., TONHOLO, J. e GOULART, M.O.F.. Eletrochemical aspects of the reduction of biologically active 2-hydroxy-3-alkyl-1,4-naphthoquinones – J. Electroanal. Chem., 507, p275-86, 2001. FERREIRA, C.A., FERREIRA, V.F., PINTO, A.V., LOPES, R.S.C., PINTO, M.C.R. e da SILVA, A.J.R.. 13-C NMR spectra of natural products V. Napnhtopyran and naphthofurandiones - An. Acad. Bras. Ciênc., 59 (1/2), págs.5-8, 1987. FERREIRA, S.B., GONZAGA, D.T.G., SANTOS, W.C., ARAÚJO, K.G.L., FERREIRA, V.F. - -lapachona: Sua importância em química medicinal e modificações estruturais. Rev. Virtual Quim., 2 (2), p140-60, 2010. 160 FERREIRA, S.P., SALOMÃO, K., DA SILVA, F.C., PINTO, A.V., KAISER, C.R., PINTO, A.C., FERREIRA, V.F. e DE CASTRO, S.L. – Synthesis and anti- Trypanosoma cruzi activity of -lapachona analogues, Eur. J. Med. Chem., 46, p3071-7, 2011. FERREIRA, V.F., JORGUEIRA, A., SOUZA, A.M.T., SILVA, M.N., SOUZA, M.C.B.V., GOUVÊA, R.M., RODRIGUES, C.R., PINTO, A.V., CASTRO, H.C., SANTOS, D.O., ARAÚJO, H.P. E BOURGUIGNON, S.C.. Trypanocidal agents with low citotoxicity to mammalian cell line: A comparison of the theoretical and biological features of lapachone derivatives, Bioorg. Med. Chem., 14, p5459-66, 2006. FERREIRA, V.F., PINTO, A.V., PINTO, M.C.F.R. e SILVA, M.M.. Catalytic reducion in pyranonaphthoquinones systems - An. Acad. Bras. Ciênc., 59 (4), p329-33, 1987. FERREIRA, V.F., SOUZA, M.C.B.V., SILVA, M.M. e PINTO, A.V.. Processos de preparação de 6-diazo--lapachona, 5-diazo--nor-lapachona e outras 5 e 6- diazo-- lapachonas substituídas nas posições 3,4, e 3 respectivamente. Pedido de Privilégio de Patente, INPI-PI 0200273-6, depósito 31/01/2002. FIESER, L.F. e HARTWELL, J.L.. The Action of Diazomethane Derivatives and of Azides on Alpha and Beta Naphthoquinones– J. Am. Chem. Soc., 57 (8), págs.1479- 84, 1935. FIESER, L.F., BERLINER, E., BONDHUS, F.J., CHANG, F.C., DAUBEN, W.G., ETTLINGER, M.G., FAWAZ, G., FIELDS, M., FIESER, M., HEIDELBERGER, C., HEYMANN, H., SELIGMAN, A.M., VAUGHAN, W.R., WILSON, A.G., WILSON, E., WU, M. E LEFFLER, M.T., HAMLINR, K.E., HATHAWAY, R.J., MATSON, E.J., MOORE, E.E., MOORE, M.B., RAPALA, R.T. e ZAUGG, H.E. - Naphthoquinone Antimalarials. I General survey - p3151-5; b) FIESER, L.F. e RICHARDSON, A.P.. Naphthoquinone Antimalarials. II. Correlation of Structure and Activity Against P. lophurae in Ducks. p3156-65; c) FIESER, L.F.. Naphthoquinone Antimalarials. III. Diene Synthesis of 1,4-Naphthoquinones. p3165- 74; d) FIESER, L.F., BERLINER, E., BONDHUS, F.J., CHANG, F.C., DAUBEN, W.G., ETTLINGER, M.G., FAWAZ, G., FIELDS, M., HEIDELBERGER, C., HEYMANN, H., VAUGHAN, W.R., WILSON, A.G., WILSON, E., WU, M., LEFFLER, M.T., HAMLIN, K.E., MATSON, E. J., MOORE, E.E., MOORE, M.B., ZAUGG, H.E.. Naphthoquinone Antimalarials. IV-XI. Synthesis. p3174-215; e) FIESER, L.F. e FIESER, M. Naphthoquinone Antimalarials. XII. The Hooker Oxidation Reaction. p3215-22; f) FIESER, L.F., ETTLINGER, M.G. e FAWAZ, G.. Naphthoquinone Antimalarials. XV. Distribution between Organic Solvents and Aqueous Buffers p3228-32; g) FIESER, L.F.. Naphthoquinone Antimalarials. XVI. Water-Soluble Derivatives of Alcoholic and Unsaturated Compounds. p3232-7; h) FIESER, L.F.. Naphthoquinone Antimalarials. XVII. Chromic Anhydride Oxidation. p3237-44, J. Am. Chem. Soc., 70, 1948. FIESER, L. F.. The alkylation of hydroxynaphthoquinone. III. A synthesis of lapachol - J. Am. Chem. Soc., 49 (3) p857-64, 1927. FONSECA, S.G.C., BRAGA, R.M.C. e SANTANA, D.P. Lapachol – química, farmacologia e métodos de dosagem, Rev. Bras. Farmacogn., 84(1), p9-16, 2003. FONSECA, S.G.C., SILVA, L.B.L., CASTRO, R.F. e SANTANA, D.P. - Validação de metodologia analítica para doseamento de soluções de lapachol por CLAE. Quím. Nova, 27 (1), p157-9, 2004. FRANCISCO, A. I., CASELLATO, A., NEVES, A.P., CARNEIRO, J.W.M., VARGAS, M.D., VISENTIN, L.C., MAGALHÃES, A., CÂMARA, C.A., PESSOA, C., COSTA-LOTUFO, L.V., FILHO, J.D.B. E MORAES, M.O. - Novel 161 2-(R-phenyl)amino-3-(2-methylpropenyl)-[1,4]-naphthoquinones: Synthesis, Characterization, Electrochemical Behavior and Antitumor Activity, J. Braz. Chem. Soc., 21 (1), p169-78, 2010. FRANCO, C.F.J., JORDÃO, A.K., FERREIRA, V.F., PINTO, A.C., SOUZA, M.C.B.V. E RESENDE, J.A.I. E CUNHA,A.C.. Synthesis of New 2- Aminocarbohydrate-1,4-Naphthoquinone Derivatives Promoted by Ultrasonic Irradiation – J. Braz. Chem. Soc., 22 (1), p1-7, 2011. FRYDMAN, B., MARTON, L.J., SUN, J.S., NEDER, K., WITIAK, D.T., LIU, A.A., WANG, H.M., MAO, Y., WU, H.Y., SANDERS, M.M. e LIU, L.F.. Induction of DNA topoisomerase II-mediated DNA cleavage by -lapachone and related naphthoquinones, Cancer Res., 57 (4), p620-7, 1997. FU, N.Y., YUAN, Y.F., CAO, Z., WANG, S.W., WANG, J.T. e PEPPE, C.. Indium (III) bromide-catalyzed preparation of dihydropyrimidones: improved protol conditions for the Biginelli reaction. Tetrahedron Lett., 58, p4801-7, 2002. FUJIWARA, A. MORI, T., IIDA, A., UEDA, S., HANO, Y., NOMURA, T., TOKUDA, H. e NISHINO, H.. Antitumor promoting naphthoquinones from Catalpaovata, J. Nat. Prod., 61(5), p629-32, 1998. GAFNER, S., WOLFENDER, J. L., NIANGA, M. e HOSTETTMANN, K.. A naphthoquinone from Newbouldia laevis roots, Phytochemistry, 48 (1), p215-6, 1998. GAFNER, S., WOLFENDER, J. L., NIANGA, M., STOECKLI-EVANS, H. e HOSTETTMANN, K.. Antifungal and antibacterial naphthoquinones from Newbouldia laevis roots, Phytochemistry, 42 (5), p1315-20, 1996. GIGANTE, B., PRAZERES, A.O., MARCELO-CURTO, M.J., CORNÉLIS, A. e LASZLO, P.,- Mild and Selective Nitration by Claycop J. Org. Chem., 60 (11), p3445-7, 1995. GLEN, V.L., HUTSON, P.R., KEHRLI, N.J., BOOTHMAN, D.A. e WILDING, G.. Quantitation of -lapachone and 3-hydroxy--lapachone in human plasma samples by reversed-phase high-performance liquid chromatography. J. Chromatogr., B: Anal. Technol. Biomed. Life Sci., 692, p181-6, 1997. GLINIS, E., MALAMIDOU-XENIKAKI, E., SKOUROS, H. SPYROUDIS, S. e TSANAKOPOULOU, M.. Arylation of lawsone through BF3-mediated coupling of its phenyliodonium ylide with activated and aromatic aldehydes. – Tetrahedron, 66, p5786-92, 2010. GOIJMAN, S. G. e STOPANNI, O. M.. Efectos del nifurtimox, benznidazol y - lapachona sobre el metabolismo del AND, ARN y proteínas en Trypanosoma cruzi, Rev. Argent. Microbiol., 15 (4), p193-204(1), 1983. GOIJMAN, S. G. e STOPANNI, O. M.. Effects of -lapachone, a peroxide-generating quinine, on macromolecule synthesis and degradation in Trypanosoma cruzi, Arch. Biochem. Biophys., 240(1), p273-80, 1985. GORMANN, R., KALOGA, M., FERREIRA, D., MARAIS, J.P.J. e KOLODZIEJ, H. – Newbouldiosides A–C, phenylethanoid glycosides from the stem bark of Newbouldia laevis - Phytochemistry, 67, p805-11, 2006. GOMEZ, J.M., GIL, L, FERROUD, C, GATEAU-OLESKER, A., MARTIN, M.T. e MARAZANO, C.. Selective Oxidation of a Keramaphidin B Model - J. Org. Chem., 66, p4898-903, 2001. GOULART, M.O.F., CIOLETTI, A.G., FILHO, J.D.S., SIMONE, C.A., CASTELLANO, E.E., EMERY, F.S., MOURA, K.C.G., PINTO, M.C.F.R. e PINTO, A.V.. Unexpected oxidation of a substituted benzo[a]phenazine: oxidative 162 cleavage of a double bond and formation of a macrolactone, Tetrahedron Lett., 44, p3581-5, 2003. GOULART, M.O.F., ZANI, C.L., TONHOLO, J., FREITAS, L.R., ABREU, F.C., OLIVEIRA, A.B., RASLAN, D.S., STARLING, S. e CHIARI, E.. Trypanocidal activity and redox potencial of heterocyclic- and 2-hydroxy-naphthoquinones, Bioorg. Med. Chem. Lett., 15, p2043-8, 1997. GUERRA, M.O., MAZONI, A.S.B., BRANDÃO, M.A.F. e PETERS, V.M.. Toxicology of lapachol in rats: embryolethality, Rev. Bras. Biol., 61, p171-4, 2001. GUIRAUD, P., STEIMAN, R., CAMPOS-TAKAKI, G. M., SEIGLE-MURANDI, F., BUOCHBERG, M. S.. Comparison of antibacterial and antifungal of lapachol and - lapachone Planta Med., 60, p373-4, 1994. GUO, B.H., KAI, G.Y., JIN, H.B. e TANG, K.X.. Taxol synthesis, Afr. J. Biotechnol., 5 (1), p15-20, 2006. GUPTA, D., PODAR, K., TAI, T-Y., LIN, B., HIDESHIMA, T., AKIYAMA, M., LEBLANC, R., CATLEY, L., MITSIADES, N., MITSIADES, C., CHAUHAN, D., MUNSHI, N.C. e ANDERSON, K.C.. -lapachone, a novel plant product, overcomes drug resistance in human multiple myeloma cells, Exp. Hematol., 30, p711-20, 2002. HANNEDOUCHE, S., SOUCHARD, J.P., JACQUEMOND-COLLET, I. e MOULIS, C.. Molluscicidal and radical scavenging activity of quinines from the root bark of Caryopteris x clandonensis, Fitoterapia, 73, p520-2, 2002. HAUN, Y.C.. Desenvolvimento da forma farmacêutica injetável a partir da -lapachona e ensaios preliminares in vivo - dissertação de mestrado, Recife, UFPE, 2006. HERNÁNDEZ-MOLINA, R., KALININA, I., ESPARZA, P., SOKOLOV, M., GONZALEZ-PLATAS, J., ESTEVEZ-BRAUN, A. e PÉREZ-SACAU, E.. Complexes of Co (II), Ni(II) and Cu (II) with lapachol, Polyhedron, 26, p4860-4, 2007. HERNDON, W.C. e GILES, W.B.. Perturbational molecular orbital method applied to [2+4] cycloadditions: olefins plus o-quinones. Chem. Commun. (Cambridge, U.K.), p497, 1969. b) HORSPOOL, W.M. e KHANDELWAL, G.D.. Photochemical addition of 1,2-naphthoquinones to p-dioxen - Chem. Commun. (Cambridge, U.K.), p1203, 1967; c) FARID, S.. Photoreactions of o-quinones with olefins: Mechanism of the 1,4-cycloaddition, Chem. Commun. (Cambridge, U.K.), p1268, 1967; d) FARID, S. e SCHOLZ, K.-H.. Photoreactions of o-quinones with olefins: Compettition between cyclo- and R-H-addition - Chem. Commun. (Cambridge, U.K.), p412, 1968; e) CHOW, Y.L. E JOSEPH, T.C. – Mechanism of phenanthrenequinone photocycloaddition to olefins - Chem. Commun. (Cambridge, U.K.), p604, 1968, f) FARID, S., HESS, D., PFUNDT, G., SCHOLZ, K.-H. e STEFFAN, G.. Photoreactions of o-quinones with olefins: a new type of reaction leading to dioxole derivates. - Chem. Commun. (Cambridge, U.K.), p638, 1968. HILDEBERT, W., KREHER, B. e LOTTER, H.. Structure Determination of New Isomeric Naphtho[2,3-b]furan-4,9-diones from Tabebuia avellanedae by the selective-INEPT technique - Helv. Chim. Acta, 72 (4), p659-67, 1989. HOLLAND, H.L., QI, J. e MANOHARAM, T.S.. Synthesis of (S)-4-hydroxy-- lapachone and biotransformation of some 4-chromanones by Mortierella isabellina ATCC 42613 - Can. J. Chem., 73, p1399-405, 1995. HOOKER, S.C.. The Constitution of Lapachic Acid (Lapachol) and Derivatives J. Chem. Soc., 61, p611-50, 1892. 163 HOOKER, S.C.. Condensation of aldehydes with beta-hydroxy-alfa-naftoquinone. Synthesis of hydrolapachol - J. Am. Chem. Soc., 58, p1163-7, 1936a. HOOKER, S.C.. The Constitution of Lapachol and its Derivatives. Part IV. Oxidation with Potassium Permanganate. J. Am. Chem. Soc., 58, p1168-73, 1936b. HOOKER, S.C.. On the oxidation of 2-hydroxy-l,4-naphthoquinone derivatives with alkaline potassium permanganate. J. Am. Chem. Soc., 58, p1174-9, 1936c. HOOKER, S.C.. On the oxidation of 2-hydroxy-l,4-naphthoquinone derivatives with alkaline potassium permanganate. Part II. Compounds with unsaturated side chains. J. Am. Chem. Soc., 58, p1179-81, 1936d. HOOKER, S.C.. Lomatiol. Part II Its occurrence, constitution, relation to and conversion into lapachol. Also a synthesis of lapachol. J. Am. Chem. Soc., 58, p1181-90, 1936e. HOOKER, S.C.. The constitution of lapachol and its derivatives. Part V, the structure of Paternò`s Isolapachone, J. Am. Chem. Soc., 58, p1190-7, 1936f. HOOKER, S.C.. Lomatiol. Part III. Oxidation with Alkaline Potassium Permanganate1,2., J. Am. Chem. Soc., 58, p1198-201, 1936g. HOOKER, S.C.. The action of light on -hydroxy--naphthoquinone1,2. - J. Am. Chem. Soc., 58, p1212-16, 1936h. HOOKER, S.C. e STEYERMARK, A.. Conversation of ortho into para, and para into ortho quinine derivatives. Part IV. Synthesis of furan derivatives of - and - naphthoquinones., J. Am. Chem. Soc., 58, p1202-7, 1936a. HOOKER, S.C. e STEYERMARK, A.. Lomatiol. Part IV. A violet quinone from the oxidation product - J. Am. Chem. Soc., 58, p1207-11, 1936b. HOOKER, S.C. e FIESER, L.F.. Concerning Wichelhaus' di--naphthoquinone oxide. J. Am. Chem. Soc., 58, p1216-1223, 1936. HORNING, E.C. (ed.). Organic Syntheses - Coll. III, Nova Iorque: John Wiley & Sons, 1955. 902p. HORSPOOL, W.M. (ed.). CRC Handbook of Organic Photochemistry and Photobiology, 1ª ed., Nova Iorque: CRC Press, 1995. 1613p. HOUGHTON, P. J., PANDEY, R. e HAWKES. Naphthoquinones and alkaloid from roots of Newbouldia laevis, Phytochemistry, 35 (6), p1602-3, 1994. HUSSAIN, H., KROHN, K., AHMAD, V.U., MIANA, G. A. e GREEN, I. R.. Lapachol: an overview. Arkivoc (Gainesville, FL, U. S.), Special Issue Reviews and Accounts, p145-71, 2007. HWANG, J.H., KIM, D.W., JO, E.J., KIM, Y.K., JO, Y.S., PARK, J.H., YOO, S.K., PARK, M.K., KWAK, T.H., KHO, Y.L., HAN, J., CHOI, H.-S., LEE, S.-H., KIM, J.M., LEE, I.K., KYUNG, T., JANG. C., CHUNG, J., KWEON, G.R. e SHONG, M. – Pharmacological stimulation of NADH oxidation ameliorates obesity and related phenotypes in mice, Diabetes, 58, p965-74. Ipê roxo. Disponível em: <http://www.redetec.org.br/inventabrasil/iperoxo.htm>. Acesso em 15 de março de 2010. ITOH, S., MARUTA, J. e FUUZUMI, S.. Addition-cycliztion reactions of nitroalkane anions with o-quinone derivatives via electron transfer in the charge-transfer complexes. J. Chem. Soc., Perkin Trans. 2, 7, p1429-33, 1996. ITOH, S., NII, K., MURE, M. e OHSHIRO, Y. Novel addition of nitroalkanes to oquinones. Tetrahedron Lett., 28 (34), p3975-8, 1987. ITOIGAWA, M., ITO, C., TAN, C. T.-W., OKUDA, M., TOKUDA, H., NISHINO, H. e FURUKAWA, H.. Cancer chemopreventive activity of naphthoquinones and their analogs from Avicennia plants, Cancer Lett. (N. Y., NY, U. S.), 174, p135-9, 2001. 164 ITOKAWA, H., MATSUMOTO, K., MORITA, H. e TAKEIA, K.. Cytotoxic naphthoquinones from Mansoa alliacea, Phytochemistry, 31 (3), p1061-2, 1992. JEONG, S.Y., PARK, S.J., YOON, S.M., JUNG, J. WOO, H.N., YI, S.L., SONG, S.Y., PARK, H.J., KIM, C., LEE, JIN S., LEE, JUNG S., CHOI, E.K. - Systemic delivery and preclinical evaluation of Au nanoparticle containing β-lapachone for radiosensitization, J. Controlled Release, 139, p239-45, 2009. JORGUEIRA, A., GOMES, T.S., SILVA, M.N., SOUZA, M.C.B.V. e FERREIRA, V.F.. Síntese de novas quinonas metídios derivados da -lapachona catalisada por iodo, 27ª RA-SBQ e XXVI Congresso Latinoamericano de química, Salvador- BA, 2004. JORGUEIRA, A., GOUVÊA, R.M., FERREIRA, V.F., SILVA, M.N. SOUZA, M.C.B.V., ZUMA, A.A., CAVALCANTI, D.F.B., ARAÚJO, H.P., SANTOS, D.O. e BOURGUIGNON, S.C.. Oxyrane derivative of -lapachone is potent growth inhibitor of Trypanosoma cruzi epimastigote forms, Parasitol Res., 99, p429-33, 2006. JOSHI, K. C., SINGH, P. e SHARMA, M. C.. Quinones and other constituents of Markhamia platycalyx and Bignonia unguiscati, J. Nat. Prod., 48(1), pág.145, 1985. JUNIOR, A.G.B.. Projetos de sistemas cromatográficos para a extração do lapachol. 2005. Tese (doutorado). UFRJ, 150p, 2005. KAUFMAN, G.M., SMITH, J.A., STOUW, G.G.V. e SHECHTER, H. – Pyrolysis of Salts of p-Tosylhydrazones. Simple Methods for Preparing Diazo Compounds and Effecting Their Carbenic Decomposition. – J. Am. Chem. Soc. 87(4), p935-7, 1965. KHAN, R. M. e MLUNGWANA, S. M. – 5-Hydroxilapachol: a cytotoxic agent from Tectona grandis Phytochemistry, 50, p439-42, 1999. KIM, S.O., KWON, J.I., JEONG, Y.K., KIM, G.Y., KIM, N.D. e CHOI, Y.H. – Induction of Egr-1 is associated with anti-metastatic and anti-invasive ability of - lapachone in human hepatocarcinoma cells, Biosci. Biotechnol. Biochem., 71 (9), p2169-76, 2007. KONGKATHIP, N., KONGKATHIP, B., SIRIPONG, P., SANGMA, C., LUANGKAMIN, S., NIYOMDECHA, M., PATTANAPA, S., PIYAVIRIYAGUL, S. e KONGSAEREE – Potent antitumor of synthetic 1,2-naphthoquinones and 1,4- naphthoquinones – Bioorg. Med. Chem., 11, p3179-91, 2003. KRISHNAN, P. e BASTOW, K.F. – Novel mechanisms of DNA topoisomerase II inhibition by pyranonaphthoquinone derivatives – eleutherin,  lapachone and - lapachone, Biochem. Pharmacol., 60, p1367-79, 2000. LARSEN, I.K., ANDERSEN, A. e PEDERSEN, B.E. – Structures of two crystalline modifications of lapachol Acta Crystallogr., Sect. C: Cryst. Struct. Commun., C48, p2009-13, 1992. LE COUTEUR, P. e BURRESON, J. – Os botões de Napoleão – as 17 moléculas que mudaram a história, 1ª edição, Rio de Janeiro: Ed. Jorge Zahar, 2003. 344p. LEE, J.I., CHOI, D.Y., CHUNG, H.S., SEO, H.G., WOO, H.J., CHOI, B.T. e CHOI, Y.H. – -lapachone induces growth inhibition and apoptosis in bladder cancer cells by modulation of Bcl-2 family and activation of caspases, Exp. Oncol., 28, p30-5, 2006. LI, C.J., AVERBOUKH, , L. e PARDEE, A.B. – -lapachone, a novel DNA topoisomerase I inhibitor with a mode of action different from camptothecin, J. Biol. Chem., 268 (30), p22463-8, 1993b. LI, C.J., LI, Y-Z., PINTO, A.B. e PARDEE, A.B. – Potent inhibition of tumor survival in vivo by -lapachone plus taxol: Combining drugs amposes different artificial checkpoints, Proc. Natl. Acad. Sci. U.S.A., 96, p13369-74, 1999. 165 LI, C.J., WANG, C. e PARDEE, A. – Induction of apoptosis by -lapachone in human prostate cancer cells, Cancer Res., 55, p3712-5, 1995. LI, C.J., ZHANG, L. J., DEZUBE, B. J., CRUMPACKER, C. S. e PARDEE, A. B. – Three inhibitors of type 1 human immunodeficiency virus long terminal repeatdirected gene expression and virus replication, Proc. Natl. Acad. Sci. U.S.A., 90, p1839-42, 1993a. LIMA, N.M.F., CORREIA, C.S., LEON, L.L., MACHADO, G.M.C., MADEIRA, M.F., SANT’ANA, A.E.G. e GOULART, M.O.F. – Antileishmanial activity of lapachol analogues, Mem. Inst. Oswaldo Cruz, 99(7), p757-61, 2004. LIMA, N.M.F., SANTOS, A.F., PORFÍRIO, Z., GOULART, M.O.F. e SANT’ANA, A.E.G. – Toxicity of lapachol and isolapachol and their potassium salts against Biomphlaria glabata, Schitosoma mansoni cercariae, Artemia salina and Tilapia nilótica, Acta Trop., 83, p43-7, 2002. LIN, C.H., ARISTOFF, P.A., JOHNSON, P.D., MCGRATH, J.P., TINIKO, J.M. e ROBERT, A. – Benzindene prostaglandins. Synthesis of optically pure 15-deoxy-U- 68,215 and its enantiomer via a modified intramolecular Wadsworth-Emmons-Wittig reaction J. Org. Chem., 52 (25), p5594-601, 1987. LINARDI, M.C.F., OLIVEIRA, M.M. e SAMPAIO, M.R.P. – A lapachol derivative active against mouse lymphocytic leukemia P-388 – J. Med. Chem., 18(11), p1159, 1975. LIRA, A.M., ARAÚJO, A.A.S., BASÍLIO, I.D.J., SANTOS, B.L.L., SANTANA, D.P. e MACEDO, R.O. – Compatibility studies of lapachol with pharmaceutical excipients for the of topical formulations – Thermochim. Acta, 457, p1-6, 2007. LISIEUX, R., JÁCOME, R.P., OLIVEIRA, A.B., RASLAN, D., MULLER, A. e WAGNER, H. – Análise de naftoquinonas em extratos brutos de raízes de Zeyheria montana M. (bolsa-de pastor), Quím. Nova, 22 (2), p175-7, 1999. LIU, T-J., LIN, S-Y. e CHAU, Y-P. – Inhibition of poly(ADP-ribose) polymerase activation attenuates -lapachone-induced necrotic cell death in human osteosarcoma cells, Toxicol. Appl. Pharmacol., 182, p116-25, 2002. LORENZI, Harri – Árvores Brasileiras – Manual de identificação e cultivo de plantas árboreas nativas do Brasil – São Paulo: Ed. Plantarum, 1992. 382p. MACHADO, T.B., PINTO, A.V., PINTO, M.C.F.R., LEAL, I.C.R., SILVA, M.G., AMARAL, A.C.F., KUSTER, R.M., SANTOS, K.R.N. – In vitro activity of Brazilian medicinal plants, naturally occurring naphthoquinones and their analogues, against methicillin-resistant Staphylococcus aureus, Int. J. Antimicrob. Agents, 21, p279-84, 2003. MAEDA, M., MURAKAMI, M., TAKEGAMI, T., OTA, T. – Promotion or suppression of experimental metastasis of B16 melanoma cells after oral administration of lapachol, Toxicol. Appl. Pharmacol., 229, p232-8, 2008. MALTA, V.R.S., PINTO, A.V., MOLFETTA, F.A., HONÓRIO, K.M., SIMONE, C.A., PEREIRA, M.A., SANTOS, R.H.A. e SILVA, A.B.F. – The influence of eletronic and steric effects in the structure-activity relationship (SAR) study of quinines compounds with biological activity against Trypanosoma cruzi, J. Mol. Struct., 634, p271-80, 2003. MANNA, S.K., GAD, Y.P., MUKHOPADHYAY, A. e AGGARWAL, B.B. – Supression of tumor necrosis factor-activated nuclear transcription factor-kB, activator protein-1, c-Jun N-terminal kinase, and apoptosis by -lapachone – Bioch. Pharmacol., 57, p763-74, 1999. MARTÍN-NAVARRO, C.M., LÓPEZ-ARENCIBIA, A., LORENZO-MORALES, J, ORAMAS-ROYO, S., HERNÁNDEZ-MOLINA, R. ESTÉVEZ-BRAUN, A., 166 RAVELO, Á.G., VALLADARES, B. e PIÑERO, J.E. – Acanthamoeba castellanii Neff: In vitro activity against the trophozoite stage of a natural sesquiterpene and a synthetic cobalt(II)–lapachol complex, Experimental Parasitol., 126, p106-8, 2010. MARTÍNEZ, M. A., DE JIMÉNEZ, M. C. L., CASTELLANO, E. E., PIRO, O. E. e AYMONINO, P. J. - Ttwo isostructural complexes of co(ii) and zn(ii) With lapacholate,imethylformamide and water,[M(lap)2(DMF)(H2O)], J. Argent. Chem. Soc., 93, p183-93, 2005. MARUYAMA, K. e NARUTA, Y. – Syntheses of - and -lapachones and their homologues by way of photochemical side chain introduction to quinine, Chem. Lett., p847-50, 1977. MATSUMOTO, T., ICHIARA, A., YANAGLYA, M., YUZAWA, T., SANNAL, A., OIKAWA, H., SAKAMURA, S. e EUGSTER, C.H. – Two new syntheses of the pyranojuglone pigment -caryopterone, Helv. Chim. Acta, 68, p2324-31, 1985. MATTAY, J. e GRIESBECK, A. (eds.) – Photochemical key steps in Organic Synthesis, Nova Iorque, E.U.A: Ed. VHC, 1994. 350p. MELTZER, R.I., LEWIS, A.D. e FISCHMAN, A., J. Org. Chem., 1959, 24, p1763-66, cit. por Kaupp, G., em Photochemical Key Steps in Organic Synthesis, Mattay, J. e Griesbeck, A., eds., Nova Iorque, E.U.A., Ed. VCH, , 1994, p224. MENNA-BARRETO, R.F.S., BEGHINI, D.G., FERREIRA, A.T.S., PINTO, A.V., CASTRO, S.L., PERALES, J. – A proteomic analysis of the mechanism of naphthoimidazoles in Trypanosoma cruzi epimastigotes in vitro, J. Proteomics, 73, p2306-15, 2010. MENNA-BARRETO, R.F.S., CÔRREA, J.R., CASCABULHO, C.M., FERNANDES, M.C., PINTO, A.V., SOARES, M.J. e CASTRO, S.L. – Naphthoimidazoles promote different death phenotypes in Trypanosoma cruzi, Parasitology, 136, p499-510, 2009c. MENNA-BARRETO, R.F.S., CÔRREA, J.R., PINTO, A.V., SOARES, M.J. e CASTRO, S.L. – Mitochondrial disruption and DNA fragmentation in Trypanosoma cruzi induced by naphthoimidazoles synthesized from -lapachone, Parasitol Res., 101, p895-905, 2007. MENNA-BARRETO, R.F.S., GONCALVES, R.L.S., COSTA, E.M., SILVA, R.S.F., PINTO, A.V. , OLIVEIRA, M.F. e CASTRO, S.L.- The effects on Trypanosoma cruzi of novel synthetic naphthoquinones are mediated by mitochondrial dysfunction, Free Radical Bio. Med., 47, p644-53, 2009a. MENNA-BARRETO, R.F.S., HENRIQUE-PONS, A., PINTO, A.V., MORGADODIAZ, J.A., SOARES, M.J. e CASTRO, S.L. – Effect of a -lapachone-derived naphthoimidazole on Trypanosoma cruzi: identification of target organelles, J. Antimicrob. Chemother., 56, p1034, 2005. MENNA-BARRETO, R.F.S., SALOMÃO, K., DANTAS, A.P., SANTA-RITA, R.M., SOARES, M.J., BARBOSA, H.S. e CASTRO, S. - Different cell death pathways induced by drugs in Trypanosoma cruzi: An ultrastructural study, Micron, 40, p157- 68, 2009b. MI, Q., LANTVIT, D., REYES-LIM, E., CHAI, H., ZHAO, W., LEE, I-S., PERAZASA’NCHEZ, S., NGASSAPA, O., KARDONO, L.B.S., RISWAN, S., HOLLINGSHEAD, M.G., MAYO, J.G., FARNSWORTH, N.R. e CORDELL, G.A. – Evaluation of the Potential Cancer Chemotherapeutic Efficacy of Natural Product Isolates Employing in Vivo Hollow Fiber Tests, J. Nat. Prod., 65, p842-50, 2002. MIAO, X.-S., SONG, P., SAVAGE, R.E., ZHONG, C., YANG, R..-Y., KIZER, D., WU, H., VOLCKOVA, E., ASHWELL, M.A., SUPKO, J.G., HE, X. e CHAN, T.C.K. – Identification of the in vitro metabolites of 3,4-dihydro-2,2-dimethyl-2H167 naphtho[1,2-b]pyran-5,6-dione (ARQ 501; -lapachone) in whole blood, Drug Metab. Dispos., 36, p641-8, 2008. MISICO, R.I. e FORZANI, E.S. – Measurement of hydrolysis decay time constants of monoarylimino derivatives of -lapachone through cyclic voltametry, Eletrochem. Commun., 5, p449-54, 2003. MOLFETTA, F.A., BRUNI, A.T., HONÓRIO, K.M. e SILVA, A.B.F. – A structureactivity relationship of quinone compounds with trypanocidal activity, Eur. J. Med. Chem., 40, p329-38, 2005. MOURA, K.C.G., EMERY, F.S., NEVES-PINTO, C., PINTO, M.C.F.R., DANTAS, A.P., SALOMÃO, K., CASTRO, S.L., PINTO, A.V. – Trypanocidal activity of isolated naphthoquinones from Tabebuia and some heterocyclic derivatives: a review from an interdisciplinary study, J. Braz. Chem. Soc., 12 (3), p325-38, 2001. MOURA, K.C.G., SALOMÃO, K., MENNA-BARRETO, R.F.S., EMERY, F.S., PINTO, M.C.F.R., PINTO, A.V. e CASTRO, S.L.– Studies on the trypanocidal activity of semi-synthetic pyran[b-4,3]naphtha[1,2-d]imidazoles from -lapachone, Eur. J. Med. Chem., 39, p639-45, 2004. NAIR, V. e TREESA, P.M. – Hetero Diels–Alder trapping of 3-methylene-1,2,4- [3H]naphthalenetrione: an efficient protocol for the synthesis of _- and _-lapachone derivatives, Tetrahedron Lett., 42, p4549-51, 2001. NEDER, K., MARTON, L.J., LIU, L.F. e FRYDMAN, B. – Reaction of -lapachone and naphthoquinones with 2-mercatoethanol: A biomimetic model of topoisomerase II Poisoning by quinones, Cell. Mol. Biol. (Noisy-le-grand, France), 44(3), p465- 74, 1998. NETTO-FERREIRA, J.C., BERNARDES, B.O., FERREIRA, A.B.B. e MIRANDA, M.A. – Laser flash photolysis study of the triplet reactivity of b-lapachones, Photochem. Photobiol. Sci.,7, p467-73, 2008a. NETTO-FERREIRA, J.C., LHIAUBET-VALLET, V., BERNARDES, B.O., FERREIRA, A.B.B. e MIRANDA, M.A. – Characterization, reactivity and photosensitizing properties of the triplet excited state of a-lapachone, Phys. Chem. Chem. Phys.,10, p6645-52, 2008b. NETTO-FERREIRA, J.C., LHIAUBET-VALLET, V., BERNARDES, B.O., FERREIRA, A.B.B. e MIRANDA, M.A. – Photosensitizing Properties of Triplet beta-Lapachones in Acetonitrile Solution. Photochem. Photobiol., 85, p153-9, 2009. NICOLAIDES, D.M., GAUTAM, D.R., LITINAS, K.E., HADJIPAVLOU-LITINA, D.J. e FYLAKTAKIDOU, K.C.- Synthesis and evaluation of the antioxidant and anti-inflammantory activities of some benzo[1]khellactone derivatives and analogues, Eur. J. Chem., 39, p323-32, 2004. NICOLAOU, K.C., PFEFFERKORN, J.A., ROECKER, A.J., CAO, G.-Q. BARLUENGA, S. E MITCHELL, H.J. – Natural Product-like Combinatorial Libraries Based on Privileged Structures. 1. General Principles and Solid-Phase Synthesis of Benzopyrans – J. Am. Chem. Soc.,122, p9939-53, 2000. NICOLAOU, K.C., PFEFFERKORN, J.A., SCHULER, F, ROECKER, AJ, CAO, G-Q E CASIDA, JE – Combinatorial synthesis of novel and potent inhibitors of NADH:ubiquinone oxidoreductase – Chem. Biol. (Oxford, U.K.), 7, p979, 2000. OLAH, G.A., MALHOTRA, R., NARANG, S.C. – Nitration – Methods and Mechanisms, Nova Iorque, E.U.A: Wiley, 1989, p61. OLIVEIRA, C.G.T., MIRANDA, F.F., FERREIRA, V.F., FREITAS, C.C., RABELLO, R.F., CARBALLIDO, J.M. e CORRÊA, L.C.D. – Synthesis and antimicrobial 168 evaluation of 3-hydrazino-naphthoquinones as analogs of lapachol – J. Braz. Chem. Soc., 12 (3), p339-45, 2001. OLIVEIRA, E.H., MEDEIROS, G.E.A., PEPPE, C., BROWN, M.A e TUCK, D.G. – The direct electrochemichal synthesis of some metal derivatives of lapachol, Can. J. Chem., 75, p499-506, 1997. OLIVEIRA, M.F. , LEMOS, T.L.G., MATTOS, M.C., SEGUNDO, T.A., SANTIAGO, G.M.P. e BRAZ-FILHO, R. – New enamine derivatives of lapachol and biological activity, An. Acad. Bras. Ciênc., 74 (2), p211-21, 2002. OLIVEIRA, R.A.S., AZEVEDO-XIMENES, E., LUZZATI, R. e GARCIA, R.C. – The hydroxy-naphthoquinone lapachol arrest mycobacterial frowth and immunomodulates host macrophages, Int. Immunopharmacol., 10, p1463-73, 2008. OSHIMA, T e NAGAI, T – Reaction of 1-Phenyldiazoethane With Tetrachloro-p- and o-Benzoquinones - Tetrahedron Lett., 26 (43), p5317-8, 1985. OTTEN, S., e ROSAZZA, J. P. Microbial transformations of natural antitumor agents: oxidation of lapachol by Penicillium notatum – Appl. Environ. Microbiol., 35, p554-7, 1978. OTTEN, S., e ROSAZZA, J. P. – Microbial transformation of natural antitumor agents: Conversion of lapachol to dehydro-o-lapachone por Curvularia lunata, Appl. Environ. Microbiol., 38 (2), p311-3, 1979. OTTEN, S., e ROSAZZA, J. P. – Oxidative ring fission of the naphthoquinones lapachol and dichloroallyl lawsone by Penicillium notatun, J. Biol. Chem., 258 (3), p1610-13, 1983. OUGH, M., LEWIS, A., BEY, E.A., GAO, J., RITCHIE, J.M., BORNMANN, W., BOOTHMAN, D.A., OBERLEY, L.W. e CULLEN, J.J. – Efficacy of -lapachone in pancreatic cancer treatment – exploiting the novel, therapeutic target NQO1, Cancer Biol. Ther., 4, p95-102, 2005. PADWA, A. (ed.) – 1,3-Dipolar Cycloaddition Chemistry, Nova Iorque, E.U.A., vol. 1, Ed. John Wiley & Sons, 1984. 1521p. PARK, B.S., KIM, J.R., LEE, S.E., KIM, K.S., TAKEOKA, G.R., AHN, Y.J. e KIM, J.H. – Selective Growth-Inhibiting effects of compounds identified in Tabebuia impetiginosa inner bark on human intestinal bacteria – J. Agric. Food Chem., 53, p1152-7, 2005. PARK B.S., LEE, H.K., LEE, S.E., PIAO, X.L., TAREOKA, G.R., WONG, R.Y., AHN, Y.J. e KIM, J.H. – Antibacterial activity of Tabebuia impetiginosa Martius ex DC (Taheebo) against Helicobater pylori – J. Ethnopharmacol., 105, p255-62, 2006. PARK, H.J., AHN, K-J., AHN, S-D., CHOI, E., LEE, S.W., WILLIAMS, B., KIM, E.J., GRIFFIN, R., BEY, E.A., BORNMANN, W.G., GAO, J., PARK, H.J., BOOTHMAN, D.A. e SONG, C.W. – Susceptibility of cancer cells to -lapachone is enhanced by ionizing radiation, Int. J. Radiat. Oncol. Biol. Phys., 61 (1), p212-9, 2005. PARK, H.J., CHOI, E., CHOI, J., AHN, K-J., KIM, E.J., JI, I-M., KOOK, Y.H., AHN, S-D., WILLIAMS, B., GRIFFIN, R., BOOTHMAN, D.A., LEE, C.K. e SONG, C.W. – Heat-induced up-regulation of NAD(P)H: Quinone oxidoreductase potentiates anticancer effects of -lapachone, Clin. Cancer Res., 11 (24), p8866-71, 2005. PATAI, S. E RAPPOPORT, Z., eds. – Supplement F2: The Chemistry of Amino, Nitroso, Nitro and Related Groups., John Wiley & Sons, Baffins Lane, Chichester, West Sussex PO19 1UD, England, 1996. 169 PATAI, S. E RAPPOPORT, Z., eds. – The Chemistry of the Quinonoid Compounds, vol. I e II, Bath, Avon: John Wiley & Sons, 1988. PAULINO, M., ALVAREDA, E.M., DENIS, P.A., BARREIRO, E.J., SPERANDIO DA SILVA, G.M., DUBIN, M., GASTELLÚ, C., AGUILERA, S. E TAPIA, O. - Studies of trypanocidal (inhibitory) power of naphthoquinones: Evaluation of quantum chemical molecular descriptors for structureeactivity relationships, Eur. J. Med. Chem., 43, p2238-46, 2008. PERAZA-SÁNCHEZ, S. R., CHÁVEZ, D., CHAI, H.B., SHIN, Y. G., GARCÍA, R., MEJÍA, M., FAIRCHILD, C. R., LANE, K. E., MENENDEZ, A. T., FARNSWORTH, N. R., CORDELL, G. A., PEZZUTO, J. M. e KINGHORN, A. D. – Cytotoxic constituents of the roots of Ekmanianthe longiflora, J. Nat. Prod., 63(4), p492-5, 2000. PÉREZ-SACAU, E., ESTÉVEZ-BRAUN, A., RAVELO, A.G., FERRO, E.A., TOKUDA, H., MUKAINAKA, T. e NISHINO, H. – Inhibitory effects of lapachol derivatives on Epstein-Barr virus activation – Bioorg. Med. Chem., 11, p483-8, 2003. PÉREZ-SACAU, E., SOTO-DELGADO, J., ESTÉVEZ-BRAUN, A., e RAVELO, A.G. – Synthesis of 9- and 10-membered macrolactones by selective ozonolysis of 1,4-diazaphenanthrene derivatives – Tetrahedron, 61, p437–45, 2005. PERRIN, D.D., ARMAREGO, W.L.F. e PERRIN, D.R. – Purification of Laboratory Chemicals, 2ª ed., Pergamon Press, Oxford, R.U., 1980. PETTIT, G. R. E HOUGHTON, L.E. – Lapachol, Can. J. Chem., 46, p2471-2, 1968. PING, H., KARAGIANIS, G., SHANXIN, W. e WATERMAN, P.G. – -lapachone and other naphthoquinones from heartwood of Paulownia kawakamii, Biochem. Syst. Ecol., 32, p1047-9, 2004. PINK, J.J., PLANCHON, S.M., TAGLIARINO, C., VARNES, M.E., SIEGEL, D. e BOOTHMAN, D.A. – NAD(P)H:Quinone oxidoreductase activity is the principal determinant of -lapachone cytotoxicity, J. Biol. Chem., 275 (8), p5416-24, 2000a. PINK, J.J., WUERZBERGER-DAVIS, S., TAGLIARINO, C., PLANCHON, S.M., YANG, X.H., FROELICH, C.J. e BOOTHMAN, D.A. – Activation of a cysteine protease in MCF-7 and T47D breast cancer cells during -lapachone-mediated apoptosis, Exp. Cell Res., 255, p144-55, 2000b. PINTO, A.V. e CASTRO, S.L. – The trypanocidal activity of naphthoquinones: A review, Molecules, 14, p4570-90, 2009. PINTO, A. V., FERREIRA, V. F. e COUTADA, L. C. M. – Síntese de análogos de xiloidonas e lapachonas - An. Acad. Bras. Ciênc., 52 (3), p477-9, 1980. PINTO, A.V., PINTO, M.C.F.R. e AGUIAR, M.A., CAPELLA, R.S. – Transformações do lapachol em nafto-[1,2-b]-furaquinonas naturais - An. Acad. Bras. Ciênc., 54 (1), p115-20, 1982b. PINTO, A.V., PINTO, M.C.F.R. e DE OLIVEIRA, C.G.T. – Síntese das alfa- e -norlapachonas, propriedades em meio ácido e reações com N-bromosuccinimida An. Acad. Bras. Ciênc., 54 (1), págs. 107-14, 1982a. PINTO, A.V., PINTO, C.N., PINTO, M.C.F.R., EMERY, F.S., MOURA, K.C.G., CARVALHO, C.E.M. e BRIN, I.M. – Fluorescent symmetric phenazines from naphthoquinones, Heterocycles, 45 (12), p2431-6, 1997b. PINTO, A.V., PINTO, C.N., PINTO, M.C.F.R., RITA, R.S., PEZZELLA, C.A.C. e CASTRO, S.L. – Trypanocidal activity of synthetic heterocyclic derivatives of active quinones from Tabebuia sp., Arzneim. Forsh., 47 (1), p74-9, 1997a. PINTO, A.V.; PINTO, M.C.R.F.; GILBERT, B.; PELLEGRINO, J.; MELLO, R.T.; Schitosomiasis Mansoni: Blockage of cercarial skin Penetration by chemical agents: 170 I. Naphthoquinoes and derivates; Trans. Roy. Soc. Trop. Med. Hyg., 71, p133-5, 1977. PINTO, C.N., MALTA, V.R.S., PINTO, M.C.F.R., SANTOS, R.H., CASTRO, S.L. e PINTO, A.V. – A trypanocidal phenazine derived from -lapchanone, J. Med. Chem., 45, p2112-15, 2002. PINTO, CLEVERSON NEVES. Quinonas bioativas de Tabebuia: Reatividade química e pontencial na síntese de heterocíclicos. 1998. Tese (Doutorado em Química Orgânica). Universidade Federal do Rio de Janeiro, Rio de Janeiro, 1998. PINTO, M.C.R., PINTO, A.V. e DE OLIVEIRA, C.G.T. – Síntese de naftoquinonas naturais a partir do lapachol - An. Acad. Bras. Ciênc., 52 (3), págs. 481-2, 1980. PITOMBEIRA, K. – Forrageiras: cochonilha do carmim provocam perdas de 100%, acessado em 30 de junho de 2012, disponível em <http://portaldoagronegocio.com.br/con teudo.php?tit=forrageiras_cochonilha_do_carmim_provoca_perdas_de_100&id=672 65>. PLANCHON, S.M., PINK, J.J., TAGLIARINO, C., BORNMANN, W.G., VARNES, M.E., BOOTHMAN, D.A. – -lapachone-induced apoptosis in human prostate cancer cells: involvement of NQO1/xip3, Exp. Cell Res., 267, p95-106, 2001. PLANCHON, S.M., WUERZBERGER, S., FRYDMAN, B., WITIAK, D.T., HUSTON, P. , CHURCH, D.R., WILDING, G. e BOOTHMAN, D.A. – -lapachone-mediated apoptosis in human promyelocytic leukemia (HL-60) and human prostate cancer cells: A p53-independent response, Cancer Res., 55, p3706-11, 1995. PLANCHON, S.M., WUERZBERGER-DAVIS, S.M., PINK, J.J., ROBERTSON, K.A., BORNMANN, W.G. e BOOTHMAN, D.A. – Bcl-2 protects against - lapachone-mediated caspase 3 activation and apoptosis in human myeloid leukemia (HL-60) cells, Oncol. Rep., 6, p485-92, 1999. Plantas medicinais. Disponível em: <http://www.amazonialegal.com.br/textos/Plantas _Medicinais.htm>. Acesso em: 22 de novembro de 2010. PORTELA, M. P. M. e STOPPANI, A.O.M. – Redox cycling of -lapachone and related o-naphthoquinones in the presence of dihydrolipoamide and oxygen, Biochem. Pharmacol., 51, p275-83, 1996. PORTELA, M. P. M., VILLAMIL, F., PERISSINOTTI, L.J. e STOPPANI, A.O.M. – Redox cycling of o-naphthoquinones in trypanosomatids Biochem. Pharmacol., 52, p1875-82, 1996. PORTUGAL, S.M., HERRERA, J.O.M., BRINN, I.M. – Anomalous eletronic absortion lapacholi-solutions – Bull. Chem. Soc. Jpn., 70 (9), p2071-6, 1997. RANA, S., BAJAJ, A., MOUT, R. e ROTELLO, V.M. – Monolayer coated gold nanoparticles for delivery applications, Adv. Drugs Delivery Rev., 64(2), p200-16, 2012. RAO, K.V., McBRIDE, T.J. e OLESON, J.J. – Recognition and evaluation of lapachol as an antitumor agent, Cancer Res., 28, p1952-4, 1968. REINICKE, K.E., BEY, E.A., BENTLE, M.S., PINK, J.J., INGALLS, S.T., HOPPEL, C.L., MISICO, R.I., ARZAC, G.M., BURTON, G., BORNMANN, W.G., SUTTON, D., GAO, J. e BOOTHMAN, D.A. – Development of -lapachone prodrugs for therapy against human cancer cells with elevated NAD(P)H:quinone oxidoreductase 1 levels, Clin. Cancer Res., 11(8), p3055-64, 2005. RIBEIRO, C.M.R., SOUZA, P.P., FERREIRA, L.L.M.D., PINTO, L.A., ALMEIDA, L.S., e Jesus, J.G. – Ciclização do lapachol induzida por sais de tálio III, Quím. Nova, 31(4), p759-62, 2008. 171 RIBEIRO, F.W., PINTO, M.C.F.R. e PINTO, A.V. – 13C-Nuclear magnetic resonance study of 1,2- and 1,4-naphthoquinones and their derivatives – J. Braz. Chem. Soc., 1 (1), p55-7, 1990. RIBEIRO, K.A.L., DE CARVALHO, C.M., MOLINA, M.T., LIMA, E.P., LÓPEZMONTERO, E., REYS, J.R.M., DE OLIVEIRA, M.B.F., PINTO, A.V., SANTANA, A.E.G e GOULART, M.O.F. – Activities of naphtoquinones against Aedes aegypti (Linnaeus, 1762) (Diptera: Culicidae), vector of dengue and Biomphalaria glabrata (Say, 1818), intermediate host of Schitosoma mansoni – Acta Trop., 111, p44-50, 2009. RIBEIRO-RODRIGUES, R., DOS SANTOS, W.G., DE OLIVEIRA, A.B., SNIECKUS, V., ZANI, C. L. e ROMANHA, A.J. – Growth inhibitory effect of naphthoquinone derivative on Trypanosoma cruzi epimastigotes, Bioorg. Med. Chem. Lett., 5 (14), p1509-12, 1995. RODRIGUES, A.M.S., PAULA, J.E., ROBLOT, F., FOURNET, A. e ESPÍNDOLA, L.S. – Larvicidal activity of Cybistax antisyphilitica against Aedes aegypti larvae – Fitoterapia, 76, p755-7, 2005. ROSSI, T. – Peru, líder em exportação de corantes naturais de cochonilha, acessado em 30 de junho de 2012, disponível em < http://www.epochtimes.com.br/mundo/68- mundo-01/961-peru-lider-em-exportacao-de-corantes-naturais-de-cochonilha.html>. SALAS, C., TAPIA, R.A., CIUDAD, K., ARMSTRONG, V., ORELLANA, M., KEMMERLING, U., FERREIRA, J., MAYA, J.D. e MORELLO, A., Trypanosoma cruzi: Activities of lapachol and - e -lapachone derivatives against epimastigote and trypomastigote forms, Bioorg. Med. Chem., 16, p688-74, 2008. SAMAJDAR, S., BECKER, F.F. e BANIK, B.K. – Montmorillonite impregnated with bismuth nitrate: A versatile reagent for the synthesis of nitro compounds of biological significance – Arkivoc (Gainesville, FL, U. S.),viii, p22-33, 2001. SANTOS, E.V.M., CARNEIRO, J.W.M. e FERREIRA, V.F. – Quantitative structureactivity relationship in aziridinyl-1,4-naphthoquinone antimalarial: study of theoretical correlations by the PM3 method – Bioorg. Med. Chem., 12, p87-93, 2004. SANTOS, M.D.F., LITIVACK-JUNIOR, J.T., ANTUNES, R.V., SILVA, T.M.S. E CAMARA, C.A.. New Adducts of Lapachol with Primary Amines, J. Braz. Chem. Soc., 22 (4), p796-800, 2011. SAVAGE, R.E., HALL, T., BRESCIANO, K., BAILEY, J., STARACE, M. e CHAN, T.C.K. - Development and validation of a liquid chromatography–tandem mass spectrometry method for the determination of ARQ 501 (_-lapachone) in plasma and tumors from nu/nu mouse xenografts, J. Chromatogr., B: Anal. Technol. Biomed. Life Sci., 872, p148-153, 2008. SATO, V. - Arma desenvolvida em laboratório combate o envelhecimento. Revista paradoxo. Disponível em: < http://www.revistaparadoxo.com/materia.php?ido=3841 >. Acesso em: 22 de novembro 2010. SCHAFFNER-SABBA, K., SCHMIDT-RUPPIN, K.H., WEHRLI, W. SCHUERCH, A.R. e WASLEY, W.F. – -lapachone: Synthesis of derivatives and activities in tumor models - J. Med. Chem., 27 (8), p990-4, 1984. SCHMIDT, T.J., MILLER-DIENER, A. e LITWACK, G.. -lapachone, a specific competitive inhibitor of ligand binding to the glucocorticoid receptor J. Biol. Chem., 259 (15), p9536-43, 1984. SCHUERCH, A.R. e WEHRLI, W. – -lapachone, an inhibitor of oncornavirus reverse transcriptase and eukaryotic DNA polymerase-. Inhibitory effect, thiol dependency and specificity, Eur. J. Biochem., 84, p197-205, 1978. 172 SHIAH, S.G., CHUANG, S.E., CHAU, Y.P., SHEN, S.C. e KUO, M.L. – Activation of c-Jun NH2-terminal and subsequent CPP32/Yama during topoisomerase inhibitor - lapachone-induced apoptosis trough an oxidation-dependent pathway, Cancer Res., 59, p391-8, 1999. SIGMAN, M.E. CHEVIS, E.A., BROWN, A., BARBAS, J.T., DABESTANI, R. e BURCH, E.L. – Enhanced photoreactivity of acenaphthylene in water: a product and mechanism study - J. Photochem. Photobiol., A, chemistry, 94, p149-55, 1996. SILVA, ANDREA ROSANE. Adição de organometálicos a orto- e paranaftoquinonas. 2004, 203f. Dissertação (Mestrado em Ciências) – Programa de Pós- Graduação em Química Orgânica. Universidade Federal Rural do Rio de Janeiro, Seropédica, 2004. SILVA, ARI MIRANDA.. Síntese de novos naftoimidazóis derivados de betalapachona e compostos relacionados, empregando irradiação na região de microondas e reagentes suportados, e outras sínteses. 2008, 154f. Tese (Doutorado em Ciências) Programa de Pós-Graduação em Química Orgânica. Universidade Federal Rural do Rio de Janeiro, Seropédica, 2008. SILVA, A. R., SILVA, A. M., FERREIRA, A. B. B., BERNARDES, B.O. e COSTA, R. L. S.. Synthesis of imidazole derivatives from -lapachone and related compounds using microwave and supporte reagents. J. Braz. Chem. Soc., 19 (6), p1230-3, 2008. SILVA, F.C., WARDELL, J.L., WARDELL, S.M.S.V., FERREIRA, V.F., LOW, J.N. e GLIDEWELL, C. – -Stacked dimers in 6-methoxy-3,3-dimethyl-3Hbenzo[ f]chromene, and centrosymmetric dimers containing C-H(arene) hydrogen bonds in racemic 3-bromo-2,2,6,6-tetramethyl-3,4-dihydro-2H,6H-1,5-dioxatriphenylene, Acta Crystallogr., Sect. C: Cryst. Struct. Commun., C63, po568-71, 2007. SILVA, MILTON NETO. Estudo de sínteses de orto-quinonas metídios e ortodiazonaftoquinonas. 2003. 383f. Tese (Doutorado em Química Orgânica). Programa de Pós-Graduação em Química Orgânica. Universidade Federal Fluminense, Niterói, 2003. SILVA, M.N., FERREIRA, S.B., JORGUEIRA, A., SOUZA, M.C.B.V., PINTO, A.V., KAISER, C.R. e FERREIRA, V.F. – Synthesis of new carbonyl and fluoroalkyl oquinone methides from -lapachone, Tetrahedron Lett., 48, p6171-3, 2007. SILVA, M.N., FERREIRA, V.F. e SOUZA, M.C.B.V. – Um panorama atual da quimica e a farmacologia de naftoquinonas, com ênfase na -lapachona e derivados – Quím. Nova, 26 (3), p407, 2003. SILVA, M.N., SOUZA, M.C.B.V., FERRERIA, V.F., PINTO, A.V., PINTO, M.C.R.F., WARDELL, S.M.S.V. e WARDELL, J.L. – Synthesis of new aldehyde derivatives from -lapachone and nor--lapachone, Arkivoc (Gainesville, FL, U. S.), p156-68, 2003. SILVA, R.S.F., COSTA, E.M., TRINDADE, U.L.T., TEIXEIRA, D.V., PINTO, M.C.F.R., SANTOS, G.L., MALTA, V.R.S., SIMONE, C.A., PINTO, A.V., Castro, S.L. – Synthesis of naphthoquinones with activity against Trypanosoma cruzi, Eur. J. Med. Chem., 41, p526-30, 2006. SILVA, R.S.F., GUIMARÃES, T.T., TEIXEIRA, D.V., LOBATO, A.P.G., PINTO, M.C.F.R., DE SIMONE, C.A., SOARES, J.G., CIOLETTI, A.G., GOULART, M.O.F. E PINTO, A.V. - The preparation of a 10-membered ring macrolactone by selective ozonolysis and the role of the dihydropyran-substituent on the MCPBAoxidation reaction profile of β-lapachone phenazines, J. Braz. Chem. Soc., 16 (5), p1074-7, 2005. 173 SILVA, R.S.F., PINTO, M.C.F.R., GOULART, M.O.F., DE SOUZA FILHO, J.D., NEVES JR., I., LOURENÇO, M.C.S. e PINTO, A.V. - A macrolactone from benzo[a]phenazine with potent activity against Mycobacterium tuberculosis, Eur. J. Med. Chem., 44, p2334-7, 2009. SILVA, T.M.S., CAMARA, C.A., BARBOSA, T.C., SOARES, A.Z., CUNHA, L.C., PINTO, A.C. e VARGAS, M.D. – Molluscicidal activity of synthetic lapachol amino and hydrogenated derivatives, Bioorg. Med. Chem., 13, p193-6, 2005. SKOV, K.A., ADOMAT, H., FARRELL, N.P. – Radiosensitization by nickel lapachol, Int. J. Radiat. Biol., 64 (6), p707-13, 1993. SMITH, MICHAEL B. Organic Synthesis. Nova Iorque: Ed. McGraw-Hill International Editons. 1994. SOLVAY PORTUGAL. Peróxido de hidrogênio. Disponível em: < http://www.solvay.pt/ solvayinportugal/plantofpovoa/productline/processodefabricacaoperoxido/0,,1666-5- 0,00.htm >. Acesso em: 01 de fevereiro de 2008. SOUSA, J.R., SILVA, G.D.F., MIYAKOSHI, T. E CHEN, C.-L. - Constituents of the Root Wood of Austroplenckia populnea var. ovate – J. Nat. Prod., 69, p1225-7, 2006. SOUSA, M. P., MATOS, M. E. O., MATOS, F. J. A., MACHADO, M. I. L. e CRAVEIRO, A. A. – Constituintes químicos ativos de plantas medicinais brasileiras – p363-7, Fortaleza: ed. UFC / Laboratório de produtos naturais, 1991. STEINERT, J., KHALAF, H. E RIMPLER, M. – High-performance liquid separation of some naturally occurring naphthoquinones and anthraquinones – J. Chromatogr., A, 723, p206-9, 1996. STOPPANI, A.O.M. – Quimioterapia de la enfermedad de Chagas – Medicina-Buenos Aire), 59 (supl. II), p147-65, 1999. SUMMERBELL, R.K. e BERGER, D. R. – Rearrangements of -halogenated ethers II. The preparation and some reactions of 2,3-diphenyl-p-dioxene - J. Am. Chem. Soc., 81, p633-9, 1959. SUN, J.S., GEISER, A.H. e FRYDMAN, B. – A preparative synthesis of lapachol and related naphthoquinones – Tetrahedron Lett., 39, p8221, 1998. SUN, X., LI, Y., LI, W., ZHANG, B., WANG, A.J., SUN, J., MIKULE, K., JIANG, Z. e LI, C.J. – Selective induction of necrotic cell death in cancer cells by -lapachone through activation of DNA dmage response pathway, Cell Cycle, 5, p2029-35, 2006. SUTTON, D., WANG, S., NASONGKLA, N., GAO, J. e DORMIDONTOVA, E.E.- Doxorubicin and -lapachone release and interaction with micellar core materials: Experiment and Modeling – Exp. Biol. Med. (London, U.K.), 232, p1090-9, 2007. SUZUKI, M., AMANO, M., CHOI, J., PARK, H.J., WILLIAMS, B.W., ONO, K. e SONG, C.W. – Synergistic effects of radiation and -lapachone in DU-145 human prostate cancer cells in vitro, Radiat. Res., 165, p525-31, 2006. TAGLIARINO, C., PINK, J.J., DUBYAK, G.R., NIEMINEN, A-L. e BOOTHMAN, D.A. – Calcium is a key signaling molecule in -lapachone-mediated cell death, J. Biol. Chem., 276 (22), p19150-9, 2001. TAKUWA, A. – An evidence for remarkable difference of the electrophilicity between two carbonyl groups in the photoexcited 1,2-naphthoquinone. Photocycloaddition reaction of 1,2-naphthoquinones with olefins. Chem. Express, p5, 1989. TAKUWA, A., NISHIGAICHI, Y., IWAMOTO, H. e SEJIRI, T. – Photocycloaddition of 1,2-naphthoquinone with 2,3-dihydro-1,4-dioxin. Formation and photocleavage of ketooxetanes. – Chem. Express, 7 (11), p877, 1992. 174 TODRE, Z.V. – Ion-radical organic chemistry: principles and applications – 2ª Ed., Boca Raton, Ed. CRC press - Taylor & Francis group, 2009. 496p. TONHOLO, J., FREITAS, L.R., ABREU, F.C., AZEVEDO, D.C., ZANI, C.L., OLIVEIRA, A.B. e GOULART, M.O.F. – Eletrochemical properties of biologically active heterocyclic naphthoquinones, J. Braz. Chem. Soc., 9 (2), p163-9, 1998. TORSSEL, K. B. G. – Natural Product Chemistry, a mechanism and biosynthetic approach to secondary metabolism, Univ. Michigan, Ed. John Wiley & Sons, 1983, 414p. TRAN, T., SAHEBA, E., ARCERIO, A.V., CHAVEZ, V., LI, Q., MARTINEZ, L.E., PRIMM, T.P. – Quinones as antimycobacterial agents, Bioorg. Med. Chem., 12, p4809-13, 2004. TURRENS, J. F., BOVERIS, A., DOCAMPO, R. e STOPPANI, A.O.M. – Acción de -lapachona sobre la producción de anión superóxido y peróxido de hidrógeno en T. cruzi, Acta Physiol. Latinoam., 29 (2-3), p162, 1979. TWARDOWSCHY, A., FREITAS, C.S., BAGGIO, C.H. MAYER, B., DOS SANTOS, A.C., PIZZOLATI, M.G., ZACARIAS, A.A., DOS SANTOS, E.P., OTUKI, M.F., MARQUES, M.C.A. – Antiulcerogenic activity of bark extract of Tabebuia avellanedae, Lorentz ex Griseb, J. Ethnopharmacol., 118, p455-9, 2008. VANNI, A., FIORE, M., SALVIA, R., CUNDARI, E., RICORDY, R., CECCARELLI, R. e DEGRASSI, F. – DNA damage and cytotoxicity induced by -lapachone: relation to poly(ADP-ribose) polymerase inhibition, Mutat. Res., 401, p55-63, 1998. VARGAS, M.D., CÂMARA, C.A., PINTO, A.C. & ROSA, M.A. – Secondary amines and unexpected 1-aza-anthraquinones from 2-methoxylapachol - Tetrahedron, 57: p9569-74, 2001. VARGAS, M.D.(BR/RJ), PINTO, A.C.(BR/RJ), CÂMARA, C.A.(BR/PB), CARVALHO, J.E. (BR/SP) e KOHN, L.K. (BR/SP). Naftoquinonas naturais e semi-sintéticas parcialamente hidrogenadas derivadas do lapachol com atividade citotóxica e antitumoral. Int CI7, PI 0502766-7 A. 20 jun. 2005, 21 fev. 2007. Revista de Propriedade Industrial, 1885. VIANA, L.M., FREITAS, M.R., RODRIGUES, S.V. e BAUMANN, W. – Extraction of lapachol from Tabebuia avellanedae wood with supercritical CO2: an altenative of soxhlet extraction Braz. J. Chem. Eng., 20 (3), p317-25, 2003. VILLAMIL, S.F., PODESTÁ, D., PORTELA, M.D.P.M. e STOPPPANI, A. – Characterization of poly(ADP-ribose)polymerasefrom Crithidia fasciculate: enzyme inhibition by -lapachone, Mol. Biochem. Parasitol., 115, p249-56, 2001. VITTAR, N.B.R., AWRUCH, J., AZIZUDDIN, K., RIVAROLA, V. - Caspaseindependent apoptosis, in human MCF-7c3 breast cancer cells, following photodynamic therapy, with a novel water-soluble phthalocyanine, Int. J. Biochem. Cell Biol., 42, p1123-31, 2010. VOGEL, A.I. – Vogel’s – Textbook of Pratical Organic Chemistry, revisada por Furniss, B.S., Hannaford, A.J. e Smith, P.W.G., 5ª ed., Londres: Longman, R.U., 1989. VOLLHARDT, K.C. e SCHORE, N.E. – Química Orgânica – Estrutura e Função – 4ª ed., Porto Alegre: Bookman, 2004. WANG, F. SAIDEL, G.M. e GAO, J. – A mechanistic model of controlled drug release from polymer millirods Effects of excipients and complex binding – J. Controlled Release, 119, p111-20, 2007. WANG, S., MILLER,W., MILTON,J., VICKER, N., STEWART, A., CHARLTON, P. MISTRY, P., HARDICK, D. e DENNY, W.A. – Structure–Activity Relationships 175 for Analogues of the Phenazine-Based Dual Topoisomerase I/II Inhibitor XR11576 – Bioorg. Med. Chem. Lett., 12, p415, 2002. WEAST, R.C. (ed.) – Handbook of chemistry and Physics, 53ª ed., Ohio, E.U.A.: The Chemical Rubber Company, 1973. WILSON, R. M., WALSH, T.F. e WHITTLE, R. – Photooxidation of vitamin K chromenol derivates - J. Am. Chem. Soc., 104, p4162-6, 1982. WUERZBERGER, S.M., PINK, J.J., PLANCHON, S.M., BYERS, K.L., BORNMANN, W.G. e BOOTHMAN, D.A. – Induction of apoptosis in MCF-7:WS8 breast cancer cels by -lapachone, Cancer Res., 58 (9), p1876-85, 1998. YAMASHITA, M., KANEKO, M., TOKUDA, H., NISHIMURA, K., KUMEDA, Y. E IIDA, A. - Synthesis and evaluation of bioactive naphthoquinones from the Brazilian medicinal plant, Tabebuia avellanedae, Bioorg. Med. Chem., 17, p6286-91, 2009. YANG, R..-Y., KIZER, D., WU, H., VOLCKOVA, E., MIAO, X.-S., ALI, S.M., TANDOM, M., SAVAGE, R.E., CHAN, T.C.K. e ASHWELL, M.A. - Synthetic methods for the preparation of ARQ 501 (-lapachone) human blood metabolites, Bioorg. Med. Chem., 16, p5635-43, 2008. ZANI, C. L.; DE OLIVEIRA, A. B. e STARLING, S. M. – Síntese de parafuranonaftoquinonas. Uma revisão. - Quím. Nova, 17 (1), p43-52, 1994. ZARRANZ, B., JASO,A., ALDANA, I. e MONGE, A. – Synthesis and Antimycobacterial Activity of New Quinoxaline-2-carboxamide 1,4-di-N -Oxide Derivatives – Bioorg. Med. Chem., 11, p2149-56, 2003. Artigo repetido em MORENO, E., ANCIZU, S., PÉREZ-SILANES, S., TORRES, E., ALDANA, I e MONGE, A. – Eur. J. Med. Chem., 45(10), p4418-26, 2010. ZIMMERMANN, M, WEHRLI, W e SCHMIDTRUPPIN, KH – Alkylidenedioxy compounds, Pedido de patente EP 0,184,990,A1, 1985.por
dc.subject.cnpqQuímicapor
dc.thumbnail.urlhttps://tede.ufrrj.br/retrieve/54364/2012%20-%20Bauer%20de%20Oliveira%20Bernardes.pdf.jpg*
dc.thumbnail.urlhttps://tede.ufrrj.br/retrieve/63744/2012%20-%20Bauer%20de%20Oliveira%20Bernardes%20%28Parte%201%20-%20Principal%29.pdf.jpg*
dc.thumbnail.urlhttps://tede.ufrrj.br/retrieve/63746/2012%20-%20Bauer%20de%20Oliveira%20Bernardes%20%28Parte%202%20-%20Anexos%29.pdf.jpg*
dc.originais.urihttps://tede.ufrrj.br/jspui/handle/jspui/3530
dc.originais.provenanceSubmitted by Sandra Pereira (srpereira@ufrrj.br) on 2020-05-18T14:30:42Z No. of bitstreams: 1 2012 - Bauer de Oliveira Bernardes.pdf: 26455317 bytes, checksum: 00a0134e16b9ac8c75310f952830c6af (MD5)eng
dc.originais.provenanceMade available in DSpace on 2020-05-18T14:30:42Z (GMT). No. of bitstreams: 1 2012 - Bauer de Oliveira Bernardes.pdf: 26455317 bytes, checksum: 00a0134e16b9ac8c75310f952830c6af (MD5) Previous issue date: 2012-11-21eng
Appears in Collections:Doutorado em Química

Se for cadastrado no RIMA, poderá receber informações por email.
Se ainda não tem uma conta, cadastre-se aqui!

Files in This Item:
File Description SizeFormat 
2012 - Bauer de Oliveira Bernardes (Parte 1 - Principal).pdfDocumento principal4.2 MBAdobe PDFThumbnail
View/Open
2012 - Bauer de Oliveira Bernardes (Parte 2 - Anexos).pdfAnexos14.45 MBAdobe PDFThumbnail
View/Open


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.