lapachone (BioDeep_00000000712)
Main id: BioDeep_00000002485
Secondary id: BioDeep_00000870847
human metabolite PANOMIX_OTCML-2023 blood metabolite Volatile Flavor Compounds
代谢物信息卡片
化学式: C15H14O3 (242.0942894)
中文名称: β拉帕醌, β-拉帕醌
谱图信息:
最多检出来源 () 0%
分子结构信息
SMILES: CC1(CCC2=C(O1)C3=CC=CC=C3C(=O)C2=O)C
InChI: InChI=1S/C15H14O3/c1-15(2)8-7-11-13(17)12(16)9-5-3-4-6-10(9)14(11)18-15/h3-6H,7-8H2,1-2H3
描述信息
Beta-lapachone is a benzochromenone that is 3,4-dihydro-2H-benzo[h]chromene-5,6-dione substituted by geminal methyl groups at position 2. Isolated from Tabebuia avellanedae, it exhibits antineoplastic and anti-inflammatory activities. It has a role as an antineoplastic agent, an anti-inflammatory agent and a plant metabolite. It is a benzochromenone and a member of orthoquinones.
Lapachone has been used in trials studying the treatment of Cancer, Carcinoma, Advanced Solid Tumors, Head and Neck Neoplasms, and Carcinoma, Squamous Cell.
beta-Lapachone is a natural product found in Markhamia stipulata, Markhamia lutea, and other organisms with data available.
Lapachone is a poorly soluble, ortho-naphthoquinone with potential antineoplastic and radiosensitizing activity. Beta-lapachone (b-lap) is bioactivated by NAD(P)H:quinone oxidoreductase-1 (NQO1), creating a futile oxidoreduction that generates high levels of superoxide. In turn, the highly reactive oxygen species (ROS) interact with DNA, thereby causing single-strand DNA breaks and calcium release from endoplasmic reticulum (ER) stores. Eventually, the extensive DNA damage causes hyperactivation of poly(ADP-ribose) polymerase-1 (PARP-1), an enzyme facilitating DNA repair, accompanied by rapid depletion of NAD+/ATP nucleotide levels. As a result, a caspase-independent and ER-stress induced mu-calpain-mediated cell death occurs in NQO1-overexpressing tumor cells. NQO1, a flavoprotein and two-electron oxidoreductase, is overexpressed in a variety of tumors.
A benzochromenone that is 3,4-dihydro-2H-benzo[h]chromene-5,6-dione substituted by geminal methyl groups at position 2. Isolated from Tabebuia avellanedae, it exhibits antineoplastic and anti-inflammatory activities.
D000890 - Anti-Infective Agents > D000998 - Antiviral Agents > D018894 - Reverse Transcriptase Inhibitors
C274 - Antineoplastic Agent > C129839 - Apoptotic Pathway-targeting Antineoplastic Agent
D004791 - Enzyme Inhibitors > D019384 - Nucleic Acid Synthesis Inhibitors
C274 - Antineoplastic Agent > C2189 - Signal Transduction Inhibitor
C274 - Antineoplastic Agent > C798 - Radiosensitizing Agent
D011838 - Radiation-Sensitizing Agents
β-Lapachone (ARQ-501;NSC-26326) is a naturally occurring O-naphthoquinone, acts as a topoisomerase I inhibitor, and induces apoptosis by inhibiting cell cycle progression.
β-Lapachone (ARQ-501;NSC-26326) is a naturally occurring O-naphthoquinone, acts as a topoisomerase I inhibitor, and induces apoptosis by inhibiting cell cycle progression.
同义名列表
48 个代谢物同义名
AfAEA centa notA inverted exclamation markAfasAA|AfAEAdaggeratrade markAfA centA centasA notA em leaderA inverted exclamation mark-Lapachone; 2H-Naphtho(1,2-b)pyran-5,6-dione, 3,4-dihydro-2,2-dimethyl-; 2H-Naphtho[1,2-b]pyran-5,6-dione, 3,4-dihydro-2,2-dimethyl-; 3,4-Dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione; 2,2-dimethyl-2H,3H,4H,5H,6H-naphtho[1,2-b]pyran-5,6-dione; 3,4-Dihydro-2,2-dimethyl-2H-naphtho[1,2-B]pyran-5,6-dione; 3,4-dihydro-2,2-dimethyl-2H-naphtho(1,2-b)pyran-5,6-dione; 2,2-Dimethyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione #; 2,2-Dimethyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione; 2,2-dimethyl-3,4-dihydrobenzo[h]chromene-5,6-dione; 2H-Naphtho[1,6-dione, 3,4-dihydro-2,2-dimethyl-; 5-17-11-00459 (Beilstein Handbook Reference); beta-Lapachone, >=98\\% (TLC); BETA LAPACHONE [WHO-DD]; Lapachone, .beta.; BCBcMAP01_000055; .beta.-Lapachone; LAPACHONE [INCI]; Lapachone, beta-; UNII-6N4FA2QQ6A; Beta -Lapachone; LAPACHONE, BETA; Beta lapachone; beta-Lapachone; Lopac0_000717; NCI60_009665; KBio2_002594; Tox21_500717; KBio3_000051; ss-Lapachone; HSCI1_000182; KBio2_000026; KBio2_005162; KBio3_000052; b-Lapachone; β-Lapachone; IDI1_033776; beta-lap-wj; Bio2_000026; Bio2_000506; ?-Lapachone; QTL1_000048; 6N4FA2QQ6A; lapachone; beta-lap; ARQ-501; NSC-26326; beta-Lapachone
数据库引用编号
14 个数据库交叉引用编号
- ChEBI: CHEBI:10429
- KEGG: C75251
- PubChem: 3885
- HMDB: HMDB0249138
- DrugBank: DB11948
- ChEMBL: CHEMBL15192
- MeSH: beta-lapachone
- ChemIDplus: 0004707328
- CAS: 4707-32-8
- medchemexpress: HY-13555
- MetaboLights: MTBLC10429
- KEGG: C10367
- PubChem: 12553
- KNApSAcK: 10429
分类词条
相关代谢途径
Reactome(0)
BioCyc(0)
PlantCyc(0)
代谢反应
0 个相关的代谢反应过程信息。
Reactome(0)
BioCyc(0)
WikiPathways(0)
Plant Reactome(0)
INOH(0)
PlantCyc(0)
COVID-19 Disease Map(0)
PathBank(0)
PharmGKB(0)
9 个相关的物种来源信息
- 265493 - Catalpa longissima: 10.1055/S-0028-1097458
- 429699 - Handroanthus guayacan:
- 429701 - Handroanthus impetiginosus:
- 9606 - Homo sapiens: -
- 1548301 - Markhamia lutea: 10.1021/NP50037A031
- 260312 - Markhamia platycalyx: 10.1021/NP50037A031
- 1489749 - Markhamia stipulata:
- 33090 - Plants: -
- 41396 - Tectona grandis: -
在这里通过桑基图来展示出与当前的这个代谢物在我们的BioDeep知识库中具有相关联信息的其他代谢物。在这里进行关联的信息来源主要有:
- PubMed: 来源于PubMed文献库中的文献信息,我们通过自然语言数据挖掘得到的在同一篇文献中被同时提及的相关代谢物列表,这个列表按照代谢物同时出现的文献数量降序排序,取前10个代谢物作为相关研究中关联性很高的代谢物集合展示在桑基图中。
- NCBI Taxonomy: 通过文献数据挖掘,得到的代谢物物种来源信息关联。这个关联信息同样按照出现的次数降序排序,取前10个代谢物作为高关联度的代谢物集合展示在桑吉图上。
- Chemical Taxonomy: 在物质分类上处于同一个分类集合中的其他代谢物
- Chemical Reaction: 在化学反应过程中,存在为当前代谢物相关联的生化反应过程中的反应底物或者反应产物的关联代谢物信息。
点击图上的相关代谢物的名称,可以跳转到相关代谢物的信息页面。
文献列表
- Karina Motta Melo Lima, Luana França Calandrini de Azevedo, Jorge Dores Rissino, Valdicley Vieira Vale, Erica Vanessa Souza Costa, Maria Fani Dolabela, Cleusa Yoshiko Nagamachi, Julio Cesar Pieczarka. Anticancer Potential and Safety Profile of β-Lapachone In Vitro.
Molecules (Basel, Switzerland).
2024 Mar; 29(6):. doi:
10.3390/molecules29061395
. [PMID: 38543031] - Lei Zhao, Hui Miao, Mingqi Quan, Shuhao Wang, Yu Zhang, Houkun Zhou, Xianglan Zhang, Zhenhua Lin, Junjie Piao. β-Lapachone induces ferroptosis of colorectal cancer cells via NCOA4-mediated ferritinophagy by activating JNK pathway.
Chemico-biological interactions.
2024 Feb; 389(?):110866. doi:
10.1016/j.cbi.2024.110866
. [PMID: 38218311] - Ashkan Kalantary-Charvadeh, Saeed Nazari Soltan Ahmad, Somayeh Aslani, Mehdi Beyrami, Mehran Mesgari-Abbasi. β-lapachone protects against doxorubicin-induced hepatotoxicity through modulation of NAD+ /SIRT-1/FXR/p-AMPK/NF-kB and Nrf2 signaling axis.
Journal of biochemical and molecular toxicology.
2023 Oct; ?(?):e23564. doi:
10.1002/jbt.23564
. [PMID: 37867446] - Yaru Li, Meiyu Feng, Tao Guo, Zheng Wang, Yanjun Zhao. Tailored Beta-Lapachone Nanomedicines for Cancer-Specific Therapy.
Advanced healthcare materials.
2023 Mar; ?(?):e2300349. doi:
10.1002/adhm.202300349
. [PMID: 36970948] - Yu Zhang, Hao Yang, Fang-Ting Wang, Xing Peng, Hai-Yang Liu, Qing-Jiang Li, Lin-Kun An. Discovery, enantioselective synthesis of myrtucommulone E analogues as tyrosyl-DNA phosphodiesterase 2 inhibitors and their biological activities.
European journal of medicinal chemistry.
2022 Aug; 238(?):114445. doi:
10.1016/j.ejmech.2022.114445
. [PMID: 35580424] - Muhammad Furqan, Alishba Fayyaz, Farhat Firdous, Hadeeqa Raza, Aishah Bilal, Rahman Shah Zaib Saleem, Syed Shahzad-Ul-Hussan, Daijie Wang, Fadia S Youssef, Nawal M Al Musayeib, Mohamed L Ashour, Hidayat Hussain, Amir Faisal. Identification and Characterization of Natural and Semisynthetic Quinones as Aurora Kinase Inhibitors.
Journal of natural products.
2022 06; 85(6):1503-1513. doi:
10.1021/acs.jnatprod.1c01222
. [PMID: 35687347] - Larissa Franca, Milena Ferraz, Maria Clara Barros, Victor Gibson, Francisco Humberto Xavier-Júnior, Nereide Stela Santos Magalhães, Mariane Lira-Nogueira. ConA-Coated Liposomes as a System to Delivery β-Lapachone to Breast Cancer Cells.
Anti-cancer agents in medicinal chemistry.
2022; 22(5):968-977. doi:
10.2174/1871520621666210624112452
. [PMID: 34170812] - Yansong Zheng, Hengce Zhang, Yueting Guo, Yuan Chen, Hanglong Chen, Yingchun Liu. X-ray repair cross-complementing protein 1 (XRCC1) loss promotes β-lapachone -induced apoptosis in pancreatic cancer cells.
BMC cancer.
2021 Nov; 21(1):1234. doi:
10.1186/s12885-021-08979-y
. [PMID: 34789190] - Bo Kyung Kim, Mi-Ri Gwon, Woo Youl Kang, In-Kyu Lee, Hae Won Lee, Sook Jin Seong, Seungil Cho, Young-Ran Yoon. Rapid Interference-free Analysis of β-Lapachone in Clinical Samples Using Liquid Chromatography-Mass Spectrometry for a Pharmacokinetic Study in Humans.
Analytical sciences : the international journal of the Japan Society for Analytical Chemistry.
2021 Aug; 37(8):1105-1110. doi:
10.2116/analsci.20p385
. [PMID: 33390413] - Camila Luiz Gomes, Victor de Albuquerque Wanderley Sales, Camila Gomes de Melo, Rosali Maria Ferreira da Silva, Rodolfo Hideki Vicente Nishimura, Larissa Araújo Rolim, Pedro José Rolim Neto. Beta-lapachone: Natural occurrence, physicochemical properties, biological activities, toxicity and synthesis.
Phytochemistry.
2021 Jun; 186(?):112713. doi:
10.1016/j.phytochem.2021.112713
. [PMID: 33667813] - Mark J Henderson, Kathleen A Trychta, Shyh-Ming Yang, Susanne Bäck, Adam Yasgar, Emily S Wires, Carina Danchik, Xiaokang Yan, Hideaki Yano, Lei Shi, Kuo-Jen Wu, Amy Q Wang, Dingyin Tao, Gergely Zahoránszky-Kőhalmi, Xin Hu, Xin Xu, David Maloney, Alexey V Zakharov, Ganesha Rai, Fumihiko Urano, Mikko Airavaara, Oksana Gavrilova, Ajit Jadhav, Yun Wang, Anton Simeonov, Brandon K Harvey. A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome.
Cell reports.
2021 04; 35(4):109040. doi:
10.1016/j.celrep.2021.109040
. [PMID: 33910017] - Chencheng Xue, Menghuan Li, Changhuang Liu, Yanan Li, Yang Fei, Yan Hu, Kaiyong Cai, Yanli Zhao, Zhong Luo. NIR-Actuated Remote Activation of Ferroptosis in Target Tumor Cells through a Photothermally Responsive Iron-Chelated Biopolymer Nanoplatform.
Angewandte Chemie (International ed. in English).
2021 04; 60(16):8938-8947. doi:
10.1002/anie.202016872
. [PMID: 33543529] - William C Putnam, Raja Reddy Kallem, Indhumathy Subramaniyan, M Shaalan Beg, Vindhya Edpuganti. Bioanalytical method development and validation of a liquid chromatography-tandem mass spectrometry method for determination of β-lapachone in human plasma.
Journal of pharmaceutical and biomedical analysis.
2020 Sep; 188(?):113466. doi:
10.1016/j.jpba.2020.113466
. [PMID: 32668395] - Daniel Clemente Moraes, Leandro Figueira Reis de Sá, Levy Tenorio Sousa Domingos, Maria do Carmo Freire Ribeiro Pinto, Rosangela Maria de Araújo Soares, Antônio Ferreira-Pereira. Synergistic interactions between β-lapachone and fluconazole in the inhibition of CaCdr2p and CaMdr1p in Candida albicans.
Revista iberoamericana de micologia.
2020 Jul; 37(3-4):104-106. doi:
10.1016/j.riam.2020.09.002
. [PMID: 33229297] - Ana L de B Oliveira, Kely C Navegantes-Lima, Valter V S Monteiro, Lucas B G Quadros, Juliana P de Oliveira, Sávio M Dos Santos, Anna C A de A Pontes, Gilson P Dorneles, Pedro R T Romão, Luiz C R Júnior, Alaíde B de Oliveira, Marta C Monteiro. β-Lapachone Increases Survival of Septic Mice by Regulating Inflammatory and Oxidative Response.
Oxidative medicine and cellular longevity.
2020; 2020(?):8820651. doi:
10.1155/2020/8820651
. [PMID: 33381269] - Xiao Wu, Benjamin Chantemargue, Florent Di Meo, Claudie Bourgaux, David Chapron, Patrick Trouillas, Véronique Rosilio. Deciphering the Peculiar Behavior of β-Lapachone in Lipid Monolayers and Bilayers.
Langmuir : the ACS journal of surfaces and colloids.
2019 11; 35(45):14603-14615. doi:
10.1021/acs.langmuir.9b02886
. [PMID: 31619039] - Tobie D Lee, Olivia W Lee, Kyle R Brimacombe, Lu Chen, Rajarshi Guha, Sabrina Lusvarghi, Bethilehem G Tebase, Carleen Klumpp-Thomas, Robert W Robey, Suresh V Ambudkar, Min Shen, Michael M Gottesman, Matthew D Hall. A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Molecular pharmacology.
2019 11; 96(5):629-640. doi:
10.1124/mol.119.115964
. [PMID: 31515284] - Eufrânio N da Silva Júnior, Guilherme A M Jardim, Claus Jacob, Uttam Dhawa, Lutz Ackermann, Solange L de Castro. Synthesis of quinones with highlighted biological applications: A critical update on the strategies towards bioactive compounds with emphasis on lapachones.
European journal of medicinal chemistry.
2019 Oct; 179(?):863-915. doi:
10.1016/j.ejmech.2019.06.056
. [PMID: 31306817] - Hyung-Jin Kim, Wa Cao, Gi-Su Oh, SeungHoon Lee, AiHua Shen, Dipendra Khadka, Su-Bin Lee, Subham Sharma, Seon Young Kim, Seong-Kyu Choe, Tae Hwan Kwak, Jin-Man Kim, Raekil Park, Hong-Seob So. Augmentation of cellular NAD+ by NQO1 enzymatic action improves age-related hearing impairment.
Aging cell.
2019 10; 18(5):e13016. doi:
10.1111/acel.13016
. [PMID: 31353811] - Marline N'Diaye, Juliette Vergnaud-Gauduchon, Valérie Nicolas, Victor Faure, Stéphanie Denis, Sonia Abreu, Pierre Chaminade, Véronique Rosilio. Hybrid Lipid Polymer Nanoparticles for Combined Chemo- and Photodynamic Therapy.
Molecular pharmaceutics.
2019 09; 16(9):4045-4058. doi:
10.1021/acs.molpharmaceut.9b00797
. [PMID: 31361499] - Davoud Sanajou, Saeed Nazari Soltan Ahmad, Vahid Hosseini, Ashkan Kalantary-Charvadeh, Yasser Marandi, Leila Roshangar, Saman Bahrambeigi, Mehran Mesgari-Abbasi. β-Lapachone protects against doxorubicin-induced nephrotoxicity via NAD+/AMPK/NF-kB in mice.
Naunyn-Schmiedeberg's archives of pharmacology.
2019 05; 392(5):633-640. doi:
10.1007/s00210-019-01619-0
. [PMID: 30671613] - Hauke Löcken, Cinzia Clamor, Klaus Müller. Napabucasin and Related Heterocycle-Fused Naphthoquinones as STAT3 Inhibitors with Antiproliferative Activity against Cancer Cells.
Journal of natural products.
2018 07; 81(7):1636-1644. doi:
10.1021/acs.jnatprod.8b00247
. [PMID: 30003778] - Xin Li, Xiaoqian Jia, Hu Niu. Nanostructured lipid carriers co-delivering lapachone and doxorubicin for overcoming multidrug resistance in breast cancer therapy.
International journal of nanomedicine.
2018; 13(?):4107-4119. doi:
10.2147/ijn.s163929
. [PMID: 30034236] - Seokuee Kim, SeungHwan Lee, Joo-Youn Cho, Seo Hyun Yoon, In-Jin Jang, Kyung-Sang Yu. Pharmacokinetics and tolerability of MB12066, a beta-lapachone derivative targeting NAD(P)H: quinone oxidoreductase 1: two independent, double-blind, placebo-controlled, combined single and multiple ascending dose first-in-human clinical trials.
Drug design, development and therapy.
2017; 11(?):3187-3195. doi:
10.2147/dddt.s151269
. [PMID: 29158665] - Hae Won Lee, Sook Jin Seong, Boram Ohk, Woo Youl Kang, Mi-Ri Gwon, Bo Kyung Kim, Hyun-Ju Kim, Young-Ran Yoon. Pharmacokinetic and safety evaluation of MB12066, an NQO1 substrate.
Drug design, development and therapy.
2017; 11(?):2719-2725. doi:
10.2147/dddt.s142339
. [PMID: 29066863] - Victor Mangas-Sanjuan, Jorge Gutiérrez-Nieto, Magdalena Echezarreta-López, Isabel González-Álvarez, Marta González-Álvarez, Vicente-Germán Casabó, Marival Bermejo, Mariana Landin. Intestinal Permeability of β-Lapachone and Its Cyclodextrin Complexes and Physical Mixtures.
European journal of drug metabolism and pharmacokinetics.
2016 Dec; 41(6):795-806. doi:
10.1007/s13318-015-0310-5
. [PMID: 26602766] - Danielle Oliveira Dos Anjos, Eliomara Sousa Sobral Alves, Vinicius Tomaz Gonçalves, Sheila Suarez Fontes, Mateus Lima Nogueira, Ana Márcia Suarez-Fontes, João Batista Neves da Costa, Fabricio Rios-Santos, Marcos André Vannier-Santos. Effects of a novel β-lapachone derivative on Trypanosoma cruzi: Parasite death involving apoptosis, autophagy and necrosis.
International journal for parasitology. Drugs and drug resistance.
2016 12; 6(3):207-219. doi:
10.1016/j.ijpddr.2016.10.003
. [PMID: 27770751] - Tae-Won Kim, Young-Jung Kim, Hyun-Tae Kim, Se-Ra Park, Ju-Young Jung. β-Lapachone enhances Mre11-Rad50-Nbs1 complex expression in cisplatin-induced nephrotoxicity.
Pharmacological reports : PR.
2016 Feb; 68(1):27-31. doi:
10.1016/j.pharep.2015.06.007
. [PMID: 26721347] - André A Vieira, Igor R Brandão, Wagner O Valença, Carlos A de Simone, Bruno C Cavalcanti, Claudia Pessoa, Teiliane R Carneiro, Antonio L Braga, Eufrânio N da Silva. Hybrid compounds with two redox centres: modular synthesis of chalcogen-containing lapachones and studies on their antitumor activity.
European journal of medicinal chemistry.
2015 Aug; 101(?):254-65. doi:
10.1016/j.ejmech.2015.06.044
. [PMID: 26142490] - Jin Hee Kim, Se Mi Lee, Cheol Hwan Myung, Kyung Rhim Lee, Seung Min Hyun, Ji Eun Lee, Young Sun Park, Se Rim Jeon, Jong Il Park, Sung Eun Chang, Jae Sung Hwang. Melanogenesis inhibition of β-lapachone, a natural product from Tabebuia avellanedae, with effective in vivo lightening potency.
Archives of dermatological research.
2015 Apr; 307(3):229-38. doi:
10.1007/s00403-015-1543-5
. [PMID: 25663088] - Gi-Su Oh, Hyung-Jin Kim, Jae-Hyuck Choi, Aihua Shen, Seong-Kyu Choe, Anzani Karna, Seung Hoon Lee, Hyang-Jeong Jo, Sei-Hoon Yang, Tae Hwan Kwak, Chul-Ho Lee, Raekil Park, Hong-Seob So. Pharmacological activation of NQO1 increases NAD⁺ levels and attenuates cisplatin-mediated acute kidney injury in mice.
Kidney international.
2014 Mar; 85(3):547-60. doi:
10.1038/ki.2013.330
. [PMID: 24025646] - André de Lima Aires, Eulália Camelo Pessoa Azevedo Ximenes, Vanessa Xavier Barbosa, Alexandre José da Silva Góes, Valdênia Maria Oliveira Souza, Mônica Camelo Pessôa de Azevedo Albuquerque. β-Lapachone: a naphthoquinone with promising antischistosomal properties in mice.
Phytomedicine : international journal of phytotherapy and phytopharmacology.
2014 Feb; 21(3):261-7. doi:
10.1016/j.phymed.2013.08.012
. [PMID: 24090700] - Sanghee Shin, Jisoo Park, Yuwen Li, Ki Nam Min, Gyeyeong Kong, Gang Min Hur, Jin Man Kim, Minho Shong, Min-Suk Jung, Jong Kook Park, Kyeong-Hoon Jeong, Myoung Gyu Park, Tae Hwan Kwak, Derek P Brazil, Jongsun Park. β-Lapachone alleviates alcoholic fatty liver disease in rats.
Cellular signalling.
2014 Feb; 26(2):295-305. doi:
10.1016/j.cellsig.2013.11.020
. [PMID: 24269941] - Gil-Tae Gang, Jung Hwan Hwang, Yong-Hoon Kim, Jung-Ran Noh, Kyoung-Shim Kim, Jin Young Jeong, Dae Eun Choi, Kang Wook Lee, Ju-Young Jung, Minho Shong, Chul-Ho Lee. Protection of NAD(P)H:quinone oxidoreductase 1 against renal ischemia/reperfusion injury in mice.
Free radical biology & medicine.
2014 Feb; 67(?):139-49. doi:
10.1016/j.freeradbiomed.2013.10.817
. [PMID: 24189322] - Yong-Hoon Kim, Jung Hwan Hwang, Kyung-Shim Kim, Jung-Ran Noh, Gil-Tae Gang, Won Keun Oh, Kyeong-Hoon Jeong, Tae Hwan Kwak, Hueng-Sik Choi, In-Kyu Lee, Chul-Ho Lee. Enhanced activation of NAD(P)H: quinone oxidoreductase 1 attenuates spontaneous hypertension by improvement of endothelial nitric oxide synthase coupling via tumor suppressor kinase liver kinase B1/adenosine 5'-monophosphate-activated protein kinase-mediated guanosine 5'-triphosphate cyclohydrolase 1 preservation.
Journal of hypertension.
2014 Feb; 32(2):306-17. doi:
10.1097/hjh.0000000000000018
. [PMID: 24241058] - Moon Hee Jeong, Nguyen Khoi Song Tran, Tae Hwan Kwak, Byung Keon Park, Chul Soon Lee, Tae-Sik Park, Young-Hoon Lee, Woo Jin Park, Dong Kwon Yang. β-Lapachone ameliorates lipotoxic cardiomyopathy in acyl CoA synthase transgenic mice.
PloS one.
2014; 9(3):e91039. doi:
10.1371/journal.pone.0091039
. [PMID: 24614171] - Aneta Zabka, Paweł Trzaskoma, Janusz Maszewski. Dissimilar effects of β-lapachone- and hydroxyurea-induced DNA replication stress in root meristem cells of Allium cepa.
Plant physiology and biochemistry : PPB.
2013 Dec; 73(?):282-93. doi:
10.1016/j.plaphy.2013.10.001
. [PMID: 24184448] - Yong-Hoon Kim, Jung Hwan Hwang, Kyung-Shim Kim, Jung-Ran Noh, Gil-Tae Gang, Sang-Woo Kim, Seung Pil Jang, Sang-Ju Lee, Sung-Ho Her, Kyeong-Hoon Jeong, Tae Hwan Kwak, Woo Jin Park, Irina V Balyasnikova, Minho Shong, Chul-Ho Lee. NQO1 activation regulates angiotensin-converting enzyme shedding in spontaneously hypertensive rats.
Cardiovascular research.
2013 Sep; 99(4):743-50. doi:
10.1093/cvr/cvt147
. [PMID: 23749777] - María Julia Lamberti, Natalia Belén Rumie Vittar, Fernando de Carvalho da Silva, Vitor Francisco Ferreira, Viviana Alicia Rivarola. Synergistic enhancement of antitumor effect of β-Lapachone by photodynamic induction of quinone oxidoreductase (NQO1).
Phytomedicine : international journal of phytotherapy and phytopharmacology.
2013 Aug; 20(11):1007-12. doi:
10.1016/j.phymed.2013.04.018
. [PMID: 23746950] - Samuel Seoane, Patricia Díaz-Rodríguez, Juan Sendon-Lago, Rosalia Gallego, Román Pérez-Fernández, Mariana Landin. Administration of the optimized β-Lapachone-poloxamer-cyclodextrin ternary system induces apoptosis, DNA damage and reduces tumor growth in a human breast adenocarcinoma xenograft mouse model.
European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V.
2013 Aug; 84(3):497-504. doi:
10.1016/j.ejpb.2012.12.019
. [PMID: 23333901] - Marília Maria Sitônio, Carlson H R de Carvalho Júnior, Ingrid de A Campos, José Bruno Nunes Ferreira Silva, Maria do Carmo Alves de Lima, Alexandre J S Góes, Maria Bernadete Sousa Maia, Pedro J Rolim Neto, Teresinha G Silva. Anti-inflammatory and anti-arthritic activities of 3,4-dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione (β-lapachone).
Inflammation research : official journal of the European Histamine Research Society ... [et al.].
2013 Jan; 62(1):107-13. doi:
10.1007/s00011-012-0557-0
. [PMID: 23052183] - Alexandra Reichstein, Silke Vortherms, Sven Bannwitz, Jan Tentrop, Helge Prinz, Klaus Müller. Synthesis and structure-activity relationships of lapacho analogues. 1. Suppression of human keratinocyte hyperproliferation by 2-substituted naphtho[2,3-b]furan-4,9-diones, activation by enzymatic one- and two-electron reduction, and intracellular generation of superoxide.
Journal of medicinal chemistry.
2012 Aug; 55(16):7273-84. doi:
10.1021/jm3009597
. [PMID: 22845014] - Yigen Li, Wenjin Huang, Shenyuan Huang, Jiulin Du, Cheng Huang. Screening of anti-cancer agent using zebrafish: comparison with the MTT assay.
Biochemical and biophysical research communications.
2012 May; 422(1):85-90. doi:
10.1016/j.bbrc.2012.04.110
. [PMID: 22560901] - Yong-Hoon Kim, Jung Hwan Hwang, Jung-Ran Noh, Gil-Tae Gang, Surendar Tadi, Yong-Hyeon Yim, Nam Ho Jeoung, Tae Hwan Kwak, Sang-Hee Lee, Gi Ryang Kweon, Jin-Man Kim, Minho Shong, In-Kyu Lee, Chul-Ho Lee. Prevention of salt-induced renal injury by activation of NAD(P)H:quinone oxidoreductase 1, associated with NADPH oxidase.
Free radical biology & medicine.
2012 Mar; 52(5):880-8. doi:
10.1016/j.freeradbiomed.2011.12.007
. [PMID: 22227174] - Dong-Oh Moon, Chang-Hee Kang, Mun-Ock Kim, You-Jin Jeon, Jae-Dong Lee, Yung Hyun Choi, Gi-Young Kim. Beta-lapachone (LAPA) decreases cell viability and telomerase activity in leukemia cells: suppression of telomerase activity by LAPA.
Journal of medicinal food.
2010 Jun; 13(3):481-8. doi:
10.1089/jmf.2008.1219
. [PMID: 20438329] - Gladys Corrêa, Ricardo Vilela, Rubem F S Menna-Barreto, Victor Midlej, Marlene Benchimol. Cell death induction in Giardia lamblia: effect of beta-lapachone and starvation.
Parasitology international.
2009 Dec; 58(4):424-37. doi:
10.1016/j.parint.2009.08.006
. [PMID: 19703583] - Edvaldo Rodrigues de Almeida, Flávia Raquel Santos Lucena, Camilla Vila Nova Soares Silva, Wilson da Silva Costa-Junior, Jouse Bezerra Cavalcanti, Gerald Bosco Lindoso Couto, Luiz Lúcio Soares da Silva, Diógenes Luís da Mota, Alex Benício da Silveira, Samuel Daniel de Sousa Filho, Aldo Cezar Passilongo da Silva. Toxicological assessment of beta-lapachone on organs from pregnant and non-pregnant rats.
Phytotherapy research : PTR.
2009 Sep; 23(9):1276-80. doi:
10.1002/ptr.2646
. [PMID: 19197915] - Mitsuaki Yamashita, Masafumi Kaneko, Harukuni Tokuda, Katsumi Nishimura, Yuko Kumeda, Akira Iida. Synthesis and evaluation of bioactive naphthoquinones from the Brazilian medicinal plant, Tabebuia avellanedae.
Bioorganic & medicinal chemistry.
2009 Sep; 17(17):6286-91. doi:
10.1016/j.bmc.2009.07.039
. [PMID: 19674905] - Neila C de Sousa, Alexandre A A de Rezende, Regildo M G da Silva, Zaira R Guterres, Ulrich Graf, Warwick E Kerr, Mário A Spanó. Modulatory effects of Tabebuia impetiginosa (Lamiales, Bignoniaceae) on doxorubicin-induced somatic mutation and recombination in Drosophila melanogaster.
Genetics and molecular biology.
2009 Apr; 32(2):382-8. doi:
10.1590/s1415-47572009005000042
. [PMID: 21637695] - Xiu-Sheng Miao, Caiyun Zhong, Yunxia Wang, Ronald E Savage, Rui-Yang Yang, Darin Kizer, Erika Volckova, Mark A Ashwell, Thomas C K Chan. In vitro metabolism of beta-lapachone (ARQ 501) in mammalian hepatocytes and cultured human cells.
Rapid communications in mass spectrometry : RCM.
2009 Jan; 23(1):12-22. doi:
10.1002/rcm.3835
. [PMID: 19051226] - Kenneth O Eyong, Ponminor S Kumar, Victor Kuete, Gabriel N Folefoc, Ephriam A Nkengfack, Sundarababu Baskaran. Semisynthesis and antitumoral activity of 2-acetylfuranonaphthoquinone and other naphthoquinone derivatives from lapachol.
Bioorganic & medicinal chemistry letters.
2008 Oct; 18(20):5387-90. doi:
10.1016/j.bmcl.2008.09.053
. [PMID: 18829316] - R E Savage, T Hall, K Bresciano, J Bailey, M Starace, T C K Chan. Development and validation of a liquid chromatography-tandem mass spectrometry method for the determination of ARQ 501 (beta-lapachone) in plasma and tumors from nu/nu mouse xenografts.
Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.
2008 Sep; 872(1-2):148-53. doi:
10.1016/j.jchromb.2008.07.031
. [PMID: 18706870] - Mary L S Queiroz, Marize C Valadares, Cristiane O Torello, Aline L Ramos, Alaíde B Oliveira, Fabíola D Rocha, Vanessa A Arruda, Walter R Accorci. Comparative studies of the effects of Tabebuia avellanedae bark extract and beta-lapachone on the hematopoietic response of tumour-bearing mice.
Journal of ethnopharmacology.
2008 May; 117(2):228-35. doi:
10.1016/j.jep.2008.01.034
. [PMID: 18343063] - Xiu-Sheng Miao, Pengfei Song, Ronald E Savage, Caiyun Zhong, Rui-Yang Yang, Darin Kizer, Hui Wu, Erika Volckova, Mark A Ashwell, Jeffrey G Supko, Xiaoying He, Thomas C K Chan. Identification of the in vitro metabolites of 3,4-dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione (ARQ 501; beta-lapachone) in whole blood.
Drug metabolism and disposition: the biological fate of chemicals.
2008 Apr; 36(4):641-8. doi:
10.1124/dmd.107.018572
. [PMID: 18180274] - Ronald E Savage, Andrew N Tyler, Xiu-Sheng Miao, Thomas C K Chan. Identification of a novel glucosylsulfate conjugate as a metabolite of 3,4-dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione (ARQ 501, beta-lapachone) in mammals.
Drug metabolism and disposition: the biological fate of chemicals.
2008 Apr; 36(4):753-8. doi:
10.1124/dmd.107.018655
. [PMID: 18227145] - Mitsuaki Yamashita, Masafumi Kaneko, Akira Iida, Harukuni Tokuda, Katsumi Nishimura. Stereoselective synthesis and cytotoxicity of a cancer chemopreventive naphthoquinone from Tabebuia avellanedae.
Bioorganic & medicinal chemistry letters.
2007 Dec; 17(23):6417-20. doi:
10.1016/j.bmcl.2007.10.005
. [PMID: 17950604] - Hyun Joo Woo, Yung Hyun Choi. Growth inhibition of A549 human lung carcinoma cells by beta-lapachone through induction of apoptosis and inhibition of telomerase activity.
International journal of oncology.
2005 Apr; 26(4):1017-23. doi:
. [PMID: 15753997]
- Huei-Ping Tzeng, Feng-Ming Ho, Kuo-Fang Chao, Min-Liang Kuo, Shoei-Yn Lin-Shiau, Shing-Hwa Liu. beta-Lapachone reduces endotoxin-induced macrophage activation and lung edema and mortality.
American journal of respiratory and critical care medicine.
2003 Jul; 168(1):85-91. doi:
10.1164/rccm.200209-1051oc
. [PMID: 12724123] - Deepak Gupta, Klaus Podar, Yu Tzu Tai, Boris Lin, Teru Hideshima, Masaharu Akiyama, Richard LeBlanc, Laurence Catley, Nicholas Mitsiades, Constantine Mitsiades, Dharminder Chauhan, Nikhil C Munshi, Kenneth C Anderson. beta-lapachone, a novel plant product, overcomes drug resistance in human multiple myeloma cells.
Experimental hematology.
2002 Jul; 30(7):711-20. doi:
10.1016/s0301-472x(02)00839-1
. [PMID: 12135668] - Y Li, C J Li, D Yu, A B Pardee. Potent induction of apoptosis by beta-lapachone in human multiple myeloma cell lines and patient cells.
Molecular medicine (Cambridge, Mass.).
2000 Dec; 6(12):1008-15. doi:
. [PMID: 11474117]
- K Müller, A Sellmer, W Wiegrebe. Potential antipsoriatic agents: lapacho compounds as potent inhibitors of HaCaT cell growth.
Journal of natural products.
1999 Aug; 62(8):1134-6. doi:
10.1021/np990139r
. [PMID: 10479319] - S H Liu, H P Tzeng, M L Kuo, S Y Lin-Shiau. Inhibition of inducible nitric oxide synthase by beta-lapachone in rat alveolar macrophages and aorta.
British journal of pharmacology.
1999 Feb; 126(3):746-50. doi:
10.1038/sj.bjp.0702341
. [PMID: 10188987] - C C Lai, T J Liu, L K Ho, M J Don, Y P Chau. beta-Lapachone induced cell death in human hepatoma (HepA2) cells.
Histology and histopathology.
1998 01; 13(1):89-97. doi:
10.14670/hh-13.89
. [PMID: 9476638] - V L Glen, P R Hutson, N J Kehrli, D A Boothman, G Wilding. Quantitation of beta-lapachone and 3-hydroxy-beta-lapachone in human plasma samples by reversed-phase high-performance liquid chromatography.
Journal of chromatography. B, Biomedical sciences and applications.
1997 Apr; 692(1):181-6. doi:
10.1016/s0378-4347(96)00497-5
. [PMID: 9187398] - C J Li, C Wang, A B Pardee. Induction of apoptosis by beta-lapachone in human prostate cancer cells.
Cancer research.
1995 Sep; 55(17):3712-5. doi:
. [PMID: 7641181]
- C J Li, L Averboukh, A B Pardee. beta-Lapachone, a novel DNA topoisomerase I inhibitor with a mode of action different from camptothecin.
The Journal of biological chemistry.
1993 Oct; 268(30):22463-8. doi:
. [PMID: 8226754]
- M Dubin, S H Fernandez Villamil, A O Stoppani. Inhibition of microsomal lipid peroxidation and cytochrome P-450-catalyzed reactions by beta-lapachone and related naphthoquinones.
Biochemical pharmacology.
1990 Apr; 39(7):1151-60. doi:
10.1016/0006-2952(90)90256-k
. [PMID: 2157443] - R Docampo, A E Vercesi. Characteristics of Ca2+ transport by Trypanosoma cruzi mitochondria in situ.
Archives of biochemistry and biophysics.
1989 Jul; 272(1):122-9. doi:
10.1016/0003-9861(89)90202-6
. [PMID: 2500059] - D A Boothman, A B Pardee. Inhibition of radiation-induced neoplastic transformation by beta-lapachone.
Proceedings of the National Academy of Sciences of the United States of America.
1989 Jul; 86(13):4963-7. doi:
10.1073/pnas.86.13.4963
. [PMID: 2740334] - T J Schmidt, A Miller-Diener, G Litwack. Beta-lapachone, a specific competitive inhibitor of ligand binding to the glucocorticoid receptor.
The Journal of biological chemistry.
1984 Aug; 259(15):9536-43. doi:
. [PMID: 6746659]
- T J Schmidt, F J Bollum, G Litwack. Correlations between the activities of DNA polymerase alpha and the glucocorticoid receptor.
Proceedings of the National Academy of Sciences of the United States of America.
1982 Aug; 79(15):4555-9. doi:
10.1073/pnas.79.15.4555
. [PMID: 6812051] - J N Lopes, F S Cruz, R Docampo, M E Vasconcellos, M C Sampaio, A V Pinto, B Gilbert. In vitro and in vivo evaluation of the toxicity of 1,4-naphthoquinone and 1,2-naphthoquinone derivatives against Trypanosoma cruzi.
Annals of tropical medicine and parasitology.
1978 Dec; 72(6):523-31. doi:
10.1080/00034983.1978.11719356
. [PMID: 367298] - R Docampo, F S Cruz, A Boveris, R P Muniz, D M Esquivel. Lipid peroxidation and the generation of free radicals, superoxide anion, and hydrogen peroxide in beta-lapachone-treated Trypanosoma cruzi epimastigotes.
Archives of biochemistry and biophysics.
1978 Mar; 186(2):292-7. doi:
10.1016/0003-9861(78)90438-1
. [PMID: 205176]