Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
J Nanobiotechnology ; 20(1): 42, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35062959

RESUMEN

Despite considerable progress has been achieved in hypoxia-associated anti-tumor therapy, the efficacy of utilizing hypoxia-activated prodrugs alone is not satisfied owing to the inadequate hypoxia within the tumor regions. In this work, a mitochondrial targeted nanoplatform integrating photodynamic therapy, photothermal therapy and hypoxia-activated chemotherapy has been developed to synergistically treat cancer and maximize the therapeutic window. Polydopamine coated hollow copper sulfide nanoparticles were used as the photothermal nanoagents and thermosensitive drug carriers for loading the hypoxia-activated prodrug, TH302, in our study. Chlorin e6 (Ce6) and triphenyl phosphonium (TPP) were conjugated onto the surface of the nanoplatform. Under the action of TPP, the obtained nanoplatform preferentially accumulated in mitochondria to restore the drug activity and avoid drug resistance. Using 660 nm laser to excite Ce6 can generate ROS and simultaneously exacerbate the cellular hypoxia. While under the irradiation of 808 nm laser, the nanoplatform produced local heat which can increase the release of TH302 in tumor cells, ablate cancer cells as well as intensify the tumor hypoxia levels. The aggravated tumor hypoxia then significantly boosted the anti-tumor efficiency of TH302. Both in vitro and in vivo studies demonstrated the greatly improved anti-cancer activity compared to conventional hypoxia-associated chemotherapy. This work highlights the potential of using a combination of hypoxia-activated prodrugs plus phototherapy for synergistic cancer treatment.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Mitocondrias/metabolismo , Nanopartículas/química , Fotoquimioterapia/métodos , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos C57BL , Nitroimidazoles/química , Nitroimidazoles/farmacocinética , Nitroimidazoles/farmacología , Mostazas de Fosforamida/química , Mostazas de Fosforamida/farmacocinética , Mostazas de Fosforamida/farmacología , Profármacos/química , Profármacos/farmacología , Distribución Tisular
2.
J Nanobiotechnology ; 19(1): 147, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011362

RESUMEN

BACKGROUND: Triple negative breast cancer (TNBC) is an aggressive tumor with extremely high mortality that results from its lack of effective therapeutic targets. As an adhesion molecule related to tumorigenesis and tumor metastasis, cluster of differentiation-44 (also known as CD44) is overexpressed in TNBC. Moreover, CD44 can be effectively targeted by a specific hyaluronic acid analog, namely, chitosan oligosaccharide (CO). In this study, a CO-coated liposome was designed, with Photochlor (HPPH) as the 660 nm light mediated photosensitizer and evofosfamide (also known as TH302) as the hypoxia-activated prodrug. The obtained liposomes can help diagnose TNBC by fluorescence imaging and produce antitumor therapy by synergetic photodynamic therapy (PDT) and chemotherapy. RESULTS: Compared with the nontargeted liposomes, the targeted liposomes exhibited good biocompatibility and targeting capability in vitro; in vivo, the targeted liposomes exhibited much better fluorescence imaging capability. Additionally, liposomes loaded with HPPH and TH302 showed significantly better antitumor effects than the other monotherapy groups both in vitro and in vivo. CONCLUSION: The impressive synergistic antitumor effects, together with the superior fluorescence imaging capability, good biocompatibility and minor side effects confers the liposomes with potential for future translational research in the diagnosis and CD44-overexpressing cancer therapy, especially TNBC.


Asunto(s)
Quitosano/farmacología , Liposomas/química , Nitroimidazoles/farmacología , Oligosacáridos/farmacología , Mostazas de Fosforamida/farmacología , Fotoquimioterapia/métodos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Quitosano/química , Femenino , Humanos , Receptores de Hialuranos , Ácido Hialurónico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanomedicina , Nitroimidazoles/química , Oligosacáridos/química , Imagen Óptica , Mostazas de Fosforamida/química , Fármacos Fotosensibilizantes/química , Profármacos/química , Neoplasias de la Mama Triple Negativas/patología
3.
Anticancer Drugs ; 31(6): 617-622, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32044797

RESUMEN

Cyclophosphamide is an inert prodrug converted into 4-hydroxycyclophosphamide (OHCP) by hepatic hydroxylation. OHCP is in equilibrium with its tautomeric aldophosphamide (ALDO). From ALDO, the cytotoxic active metabolites are formed enzymatically by phosphodiesterases; these are the alkylating metabolite phosphoramide mustard (PAM) and the proapoptotic aldehyde 3-hydroxypropanal (HPA). PAM damages the DNA by alkylation; HPA amplifies the thereby induced apoptosis. The generally accepted view that acrolein, which is believed to be formed in the formation of PAM by ß-elimination from ALDO would be mainly responsible for the toxicity of cyclophosphamide, has to be revised because no acrolein is formed in the systemic circulation of patients after cyclophosphamide administration. It is shown that not acrolein, but OHCP itself is the true toxic metabolite of cyclophosphamide. Toxicity tests with OHCP and PAM were carried out, which demonstrated that OHCP unfolds its toxicity, not as a carrier of PAM but is toxic itself by reacting with nucleophilic groups of macromolecules, for example, thiol groups of membrane proteins. Further experiments demonstrate that the toxicity of oxazaphosphorine cytostatics may be drastically reduced if the formation of the pharmacologically active metabolite ALDO bypasses the formation of OHCP. Toxicity experiments in mice with S-ethanol-cyclophosphamide (SECP) that hydrolyzes to OHCP show that SECP is as toxic as OHCP, whereas the thiazolidine of ALDO, which hydrolyzes to ALDO bypassing OHCP is 7-9 times less toxic without loss of antitumor activity.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , Proliferación Celular/efectos de los fármacos , Ciclofosfamida/análogos & derivados , Ciclofosfamida/toxicidad , Leucemia P388/patología , Mostazas de Fosforamida/toxicidad , Animales , Antineoplásicos Alquilantes/química , Ciclofosfamida/química , Femenino , Leucemia P388/tratamiento farmacológico , Masculino , Ratones , Mostazas de Fosforamida/química , Pruebas de Toxicidad
4.
Bioorg Chem ; 98: 103747, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32208207

RESUMEN

The tumor microenvironment in chondrosarcoma (CHS), a chemo- and radio-resistant cancer provides unique hallmarks for developing a chondrosarcoma targeted drug-delivery system. Tumor targeting could be achieved using a quaternary ammonium function (QA) as a ligand for aggrecan, the main high negative charged proteoglycan of the extracellular matrix of CHS, and a 2-nitroimidazole as trigger that enables hypoxia-responsive drug release. In a previous work, ICF05016 was identified as efficient proteoglycan-targeting hypoxia-activated prodrug in a human extraskeletal myxoid chondrosarcoma model in mice and a first study of the structure-activity relationship of the QA function and the alkyl linker length was conducted. Here, we report the second part of the study, namely the modification of the nitro-aromatic trigger and the position of the proteoglycan-targeting ligand at the aromatic ring as well as the nature of the alkylating mustard. Synthetic approaches have been established to functionalize the 2-nitroimidazole ring at the N-1 and C-4 positions with a terminal tertiary alkyl amine, and to perform the phosphorylation step namely through the use of an amine borane complex, leading to phosphoramide and isophosphoramide mustards and also to a phosphoramide mustard bearing four 2-chloroethyl chains. In a preliminary study using a reductive chemical activation, QA-conjugates, except the 4-nitrobenzyl one, were showed to undergo efficient cleavage with release of the corresponding mustard. However N,N,N-trimethylpropylaminium tethered to the N-1 or C-4 positions of the imidazole seemed to hamper the enzymatic reduction of the prodrugs and all tested compounds featured moderate selectivity toward hypoxic cells, likely not sufficient for application as hypoxia-activated prodrugs.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Condrosarcoma/tratamiento farmacológico , Diseño de Fármacos , Neoplasias de los Tejidos Conjuntivo y Blando/tratamiento farmacológico , Mostazas de Fosforamida/farmacología , Profármacos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias Óseas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Condrosarcoma/patología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Neoplasias de los Tejidos Conjuntivo y Blando/patología , Mostazas de Fosforamida/síntesis química , Mostazas de Fosforamida/química , Profármacos/síntesis química , Profármacos/química , Relación Estructura-Actividad
5.
Molecules ; 23(8)2018 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-30082625

RESUMEN

Novel mustard functionalized sophoridine derivatives were synthesized and evaluated for their cytotoxicity against of a panel of various cancer cell lines. They were shown to be more sensitive to S180 and H22 tumor cells with IC50 values ranging from 1.01⁻3.65 µM, and distinctly were more cytotoxic to cancer cells than normal cell L929. In addition, compounds 7a, 7c, and 7e displayed moderate tumor suppression without apparent organ toxicity in vivo against mice bearing H22 liver tumors. Furthermore, they arrested tumor cells in the G1 phase and induced cellular apoptosis. Their potential binding modes with DNA-Top I complex have also been investigated.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Mostazas de Fosforamida/química , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Relación Estructura-Actividad
6.
Chem Res Toxicol ; 29(2): 190-202, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26692166

RESUMEN

N,N-Bis-(2-chloroethyl)-phosphorodiamidic acid (phosphoramide mustard, PM) and N,N-bis-(2-chloroethyl)-amine (nornitrogen mustard, NOR) are the two biologically active metabolites of cyclophosphamide, a DNA alkylating drug commonly used to treat lymphomas, breast cancer, certain brain cancers, and autoimmune diseases. PM and NOR are reactive bis-electrophiles capable of cross-linking cellular biomolecules to form covalent DNA-DNA and DNA-protein cross-links (DPCs). In the present work, a mass spectrometry-based proteomics approach was employed to characterize PM- and NOR-mediated DNA-protein cross-linking in human cells. Following treatment of human fibrosarcoma cells (HT1080) with cytotoxic concentrations of PM, over 130 proteins were found to be covalently trapped to DNA, including those involved in transcriptional regulation, RNA splicing/processing, chromatin organization, and protein transport. HPLC-ESI(+)-MS/MS analysis of proteolytic digests of DPC-containing DNA from NOR-treated cells revealed a concentration-dependent formation of N-[2-[cysteinyl]ethyl]-N-[2-(guan-7-yl)ethyl]amine (Cys-NOR-N7G) conjugates, confirming that it cross-links cysteine thiols of proteins to the N7 position of guanines in DNA. Cys-NOR-N7G adduct numbers were higher in NER-deficient xeroderma pigmentosum cells (XPA) as compared with repair proficient cells. Furthermore, both XPA and FANCD2 deficient cells were sensitized to PM treatment as compared to that of wild type cells, suggesting that Fanconi anemia and nucleotide excision repair pathways are involved in the removal of cyclophosphamide-induced DNA damage.


Asunto(s)
Alquilantes/química , ADN/química , Compuestos de Mostaza Nitrogenada/química , Mostazas de Fosforamida/química , Proteínas/química , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , ADN/metabolismo , Aductos de ADN/análisis , Humanos , Péptidos/análisis , Proteínas/metabolismo , Proteómica , Espectrometría de Masa por Ionización de Electrospray
7.
Bioorg Med Chem ; 24(12): 2697-706, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27156193

RESUMEN

A series of Glutaryl-Hyp-Ala-Ser-Chg-Gln-4-aminobenzyl phosphoramide mustard conjugates (1a-e) was designed and synthesized as potential prodrugs for site-specific activation by PSA in prostate cancer cells. All conjugates were found to be substrates of PSA with cleavage occurring between Gln and the para-aminobenzyl (PAB) linker. Structure-activity relationship studies on these conjugates indicated that introduction of electron-withdrawing fluorine(s) on the phenyl ring in the PAB linker uniformly improved the chemical stability of the conjugates while the position of substitution affected differently the self-immolative process of conjugates upon proteolysis. Introduction of a fluorine at ortho position to benzylic phosphoramide as in 1b results in better stability of the conjugate prior to activation while maintaining its antiproliferative activity upon activation by PSA. The conjugate 1b with 2-fluoro substitution was identified as a promising lead for further evaluation and optimization in the development of prostate cancer-targeted prodrugs.


Asunto(s)
Antineoplásicos/química , Diseño de Fármacos , Péptidos/química , Mostazas de Fosforamida/química , Profármacos/química , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Péptidos/síntesis química , Péptidos/metabolismo , Péptidos/farmacología , Mostazas de Fosforamida/síntesis química , Mostazas de Fosforamida/metabolismo , Mostazas de Fosforamida/farmacología , Profármacos/síntesis química , Profármacos/metabolismo , Profármacos/farmacología , Neoplasias de la Próstata/metabolismo , Relación Estructura-Actividad
8.
Bioorg Med Chem ; 22(21): 5747-56, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25440502

RESUMEN

A series of bis(sulfonylethyl) and mono(sulfonylethyl) phenyl phosphotriesters of zidovudine (3'-azido-3'-deoxythymidine, AZT) were synthesized as potential anticancer prodrugs that liberate AZT monophosphate via nonenzymatic ß-elimination mechanism. Stability studies demonstrated that all the synthesized prodrugs spontaneously liberate AZT monophosphate with half-lives in the range of 0.07-278.8h under model physiological conditions in 0.1M phosphate buffer at pH 7.4 and 37 °C. Analogous to aldophosphamide, the elimination rates were accelerated in the presence of reconstituted human plasma under the same conditions. Among the compounds, 3, 4, 8, and 10 were comparable or superior to AZT against five established human cancerous cell lines in vitro. Moreover, the selected compounds were equally sensitive to both the wild-type osteosarcoma 143 B and the thymidine kinase-deficient 143 B/TK(-) cell lines. The findings are consistent with that these compounds deliver AZT monophosphate intracellularly.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Profármacos/síntesis química , Profármacos/farmacología , Sulfonas/química , Zidovudina/química , Zidovudina/farmacología , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ésteres , Semivida , Humanos , Células MCF-7 , Mostazas de Fosforamida/química , Profármacos/farmacocinética , Zidovudina/farmacocinética
9.
Chin J Cancer ; 33(2): 80-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23845143

RESUMEN

Hypoxia, a state of low oxygen, is a common feature of solid tumors and is associated with disease progression as well as resistance to radiotherapy and certain chemotherapeutic drugs. Hypoxic regions in tumors, therefore, represent attractive targets for cancer therapy. To date, five distinct classes of bioreactive prodrugs have been developed to target hypoxic cells in solid tumors. These hypoxia-activated prodrugs, including nitro compounds, N-oxides, quinones, and metal complexes, generally share a common mechanism of activation whereby they are reduced by intracellular oxidoreductases in an oxygen-sensitive manner to form cytotoxins. Several examples including PR-104, TH-302, and EO9 are currently undergoing phase II and phase III clinical evaluation. In this review, we discuss the nature of tumor hypoxia as a therapeutic target, focusing on the development of bioreductive prodrugs. We also describe the current knowledge of how each prodrug class is activated and detail the clinical progress of leading examples.


Asunto(s)
Antineoplásicos/farmacología , Hipoxia de la Célula/efectos de los fármacos , Neoplasias , Profármacos/farmacología , Antraquinonas/química , Antraquinonas/farmacología , Antineoplásicos/química , Aziridinas/química , Aziridinas/farmacología , Humanos , Indolquinonas/química , Indolquinonas/farmacología , Estructura Molecular , NAD(P)H Deshidrogenasa (Quinona)/química , NAD(P)H Deshidrogenasa (Quinona)/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Compuestos de Mostaza Nitrogenada/química , Compuestos de Mostaza Nitrogenada/farmacología , Nitroimidazoles/química , Nitroimidazoles/farmacología , Mostazas de Fosforamida/química , Mostazas de Fosforamida/farmacología , Profármacos/química , Tirapazamina , Triazinas/química , Triazinas/farmacología
10.
Bioorg Med Chem ; 21(23): 7507-14, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24139844

RESUMEN

In our continued effort to develop prodrugs of phosphoramide mustard, conjugates of 4-aminocyclophosphamide (4-NH2-CPA) with three PSA-specific peptides were synthesized and evaluated as substrates of PSA. These include conjugates of cis-(2R,4R)-4-NH2-CPA with a tetrapeptide Succinyl-Ser-Lys-Leu-Gln-OH, a hexapeptide Succinyl-His-Ser-Ser-Lys-Leu-Gln-OH, and a pentapeptide Glutaryl-Hyp-Ala-Ser-Chg-Gln-OH. These conjugates were cleaved by PSA efficiently and exclusively after the expected glutamine residue to release 4-NH2-CPA, the activated prodrug form of phosphoramide mustard. The cleavage was most efficient for the pentapeptide conjugate 3 (Glutaryl-Hyp-Ala-Ser-Chg-Gln-NH-CPA), which showed a half-life of 55 min with PSA, followed by the hexapeptide conjugate 2 (Succinyl-His-Ser-Ser-Lys-Leu-Gln-NH-CPA) and the tertrapeptide conjugate 1 (Succinyl-Ser-Lys-Leu-Gln-NH-CPA) with half-lives of 6.5 and 12h, respectively. These results indicate a potential of the conjugate 3 as an anticancer prodrug of phosphoramide mustard for selective PSA activation.


Asunto(s)
Antineoplásicos/química , Oligopéptidos/química , Mostazas de Fosforamida/química , Profármacos/química , Antígeno Prostático Específico/metabolismo , Antineoplásicos/metabolismo , Humanos , Oligopéptidos/metabolismo , Mostazas de Fosforamida/metabolismo , Profármacos/metabolismo , Antígeno Prostático Específico/química
11.
Folia Biol (Krakow) ; 61(1-2): 31-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23767290

RESUMEN

Mafosfamide cyclohexylamine salt (D-17272), 4-hydro-peroxy-cyclophosphamide (D-18864) and glufosfamide (D-19575, beta-D-glucose-isophosphoramide mustard) are new generation oxazaphosphorine agents. The present investigation was undertaken to determine the activity of these three oxazaphosphorines in human promyelocytic leukemia HL-60 cells. The research was conducted using the spectrophotometric MTT assay and the electronic Beckman Coulter and microscopy methods. Functional and morphological changes were observed after exposure of HL-60 cells to the oxazaphosphorine agents. The various patterns of temporary alterations in cell viability, size and count, and also in the frequency of leukemic cells undergoing mitotic catastrophe, apoptosis and necrosis, were shown. Different leukemic cell responses to the action of the three oxazaphosphorines were evaluated. These are the first data comparing the in vitro activity of D-17272, D-18864 and D-19575 against human promyelocytic leukemia cells.


Asunto(s)
Antineoplásicos/farmacología , Mostazas de Fosforamida/farmacología , Antineoplásicos/química , Relación Dosis-Respuesta a Droga , Células HL-60 , Humanos , Estructura Molecular , Mostazas de Fosforamida/química
12.
Antioxid Redox Signal ; 35(11): 904-915, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-32787454

RESUMEN

Aims: In hypoxic tumor microenvironments, the strongly reducing redox environment reduces evofosfamide (TH-302) to release a cytotoxic bromo-isophosphoramide (Br-IPM) moiety. This drug therefore preferentially attacks hypoxic regions in tumors where other standard anticancer treatments such as chemotherapy and radiation therapy are often ineffective. Various combination therapies with evofosfamide have been proposed and tested in preclinical and clinical settings. However, the treatment effect of evofosfamide monotherapy on tumor hypoxia has not been fully understood, partly due to the lack of quantitative methods to assess tumor pO2in vivo. Here, we use quantitative pO2 imaging by electron paramagnetic resonance (EPR) to evaluate the change in tumor hypoxia in response to evofosfamide treatment using two pancreatic ductal adenocarcinoma xenograft models: MIA Paca-2 tumors responding to evofosfamide and Su.86.86 tumors that do not respond. Results: EPR imaging showed that oxygenation improved globally after evofosfamide treatment in hypoxic MIA Paca-2 tumors, in agreement with the ex vivo results obtained from hypoxia staining by pimonidazole and in apparent contrast to the decrease in Ktrans observed in dynamic contrast-enhanced magnetic resonance imaging (DCE MRI). Innovations: The observation that evofosfamide not only kills the hypoxic region of the tumor but also improves oxygenation in the residual tumor regions provides a rationale for combination therapies using radiation and antiproliferatives post evofosfamide for improved outcomes. Conclusion: This study suggests that reoxygenation after evofosfamide treatment is due to decreased oxygen demand rather than improved perfusion. Following the change in pO2 after treatment may therefore yield a way of monitoring treatment response. Antioxid. Redox Signal. 35, 904-915.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/terapia , Hipoxia de la Célula/efectos de los fármacos , Nitroimidazoles/farmacología , Neoplasias Pancreáticas/terapia , Mostazas de Fosforamida/farmacología , Profármacos/farmacología , Animales , Antineoplásicos/química , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Nitroimidazoles/química , Oxidación-Reducción , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Mostazas de Fosforamida/química , Profármacos/química
13.
Antimicrob Agents Chemother ; 54(3): 1193-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20028822

RESUMEN

Nitroheterocyclic prodrugs have been used to treat trypanosomal diseases for more than 40 years. Recently, the key step involved in the activation of these compounds has been elucidated and shown to be catalyzed by a type I nitroreductase (NTR). This class of enzyme is normally associated with bacteria and is absent from most eukaryotes, with trypanosomes being a major exception. Here we exploit this difference by evaluating the trypanocidal activity of a library of nitrobenzylphosphoramide mustards against bloodstream-form Trypanosoma brucei parasites. Biochemical screening against the purified enzyme revealed that a subset of halogenated nitroaromatic compounds were effective substrates for T. brucei NTR (TbNTR), having apparent K(cat)/K(m) values approximately 100 times greater than nifurtimox. When tested against T. brucei, cytotoxicity mirrored enzyme activity, with 50% inhibitory concentrations of the most potent substrates being less than 10 nM. T. brucei NTR plays a key role in parasite killing: heterozygous lines displayed resistance to the compounds, while parasites overexpressing the enzyme showed hypersensitivity. We also evaluated the cytotoxicities of substrates with the highest trypanocidal activities by using mammalian THP-1 cells. The relative toxicities of these newly identified compounds were much lower than that of nifurtimox. We conclude that halogenated nitrobenzylphosphoramide mustards represent a novel class of antitrypanosomal agents, and their efficacy validates the strategy of specifically targeting NTR activity to develop new therapeutics.


Asunto(s)
Nitrorreductasas/metabolismo , Mostazas de Fosforamida , Tripanocidas , Trypanosoma brucei brucei/efectos de los fármacos , Animales , Línea Celular , Activación Enzimática , Humanos , Oxidación-Reducción , Pruebas de Sensibilidad Parasitaria , Mostazas de Fosforamida/química , Mostazas de Fosforamida/metabolismo , Mostazas de Fosforamida/farmacología , Mostazas de Fosforamida/toxicidad , Relación Estructura-Actividad , Especificidad por Sustrato , Tripanocidas/química , Tripanocidas/metabolismo , Tripanocidas/farmacología , Tripanocidas/toxicidad , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/crecimiento & desarrollo
14.
Bioorg Med Chem Lett ; 18(14): 4059-63, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18556201

RESUMEN

N-(2,2-Dimethyl-2-(2-nitrophenyl)acetyl)-4-aminocyclophosphamide isomers (DMNA-NH-CPA, 4) were synthesized stereospecifically from Boc-L-Hse(OBn)-OH and the degradation of the corresponding reduced amine 5a was investigated by UV/vis spectroscopy and LC/MS. The rate of cyclization of 5a was found to increase with decreasing pH, with half-lives ranging from 3.2 to 54 min at pH 4-7.4, suggesting that the cyclization is catalyzed by the hydronium ions. LC/MS analysis of the degradation products of 5a indicates that 4-aminocyclophosphamide is rapidly released from 4 upon reductive activation under acidic conditions and further decomposes into the cytotoxic phosphoramide mustard. These results validated 4-aminocyclophosphamide as a prodrug form of phosphoramide mustard and suggest that compound 4 can potentially be used as a prodrug of phosphoramide mustard for bioreductive activation.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacología , Mostazas de Fosforamida/farmacología , Catálisis , Cromatografía Liquida/métodos , Diseño de Fármacos , Concentración de Iones de Hidrógeno , Iones , Cinética , Espectrometría de Masas/métodos , Modelos Químicos , Compuestos Onio/química , Mostazas de Fosforamida/química , Isoformas de Proteínas , Espectrofotometría Ultravioleta/métodos , Factores de Tiempo
15.
Biochem Pharmacol ; 156: 265-280, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30134191

RESUMEN

Evofosfamide (TH-302) is a clinical-stage hypoxia-activated prodrug with proven efficacy against hypoxic cells in preclinical tumour models. TH-302 is designed to release the DNA crosslinking agent bromo-isophosphoramide mustard (Br-IPM) when reduced in hypoxic tissue. Br-IPM is considered to diffuse locally from hypoxic regions, eliciting additional tumour cell killing, but the latter 'bystander effect' has not been demonstrated directly. Previous studies with multicellular co-cultures that included cells expressing the E. coli nitroreductase NfsA as a model TH-302 reductase have provided clear evidence of a bystander effect (which we confirm in the present study). However, NfsA is an oxygen-insensitive two-electron reductase that is not expected to generate the nitro radical intermediate that has been demonstrated to fragment to release Br-IPM. Here, we use mass spectrometry methods to characterise TH-302 metabolites generated by one-electron reduction (steady-state radiolysis by ionising radiation and cellular metabolism under hypoxia, including HCT116 cells that overexpress P450 oxidoreductase, POR) or by NfsA expressed in HCT116 cells under oxic conditions, and investigate the stability and cytotoxicity of these products. Br-IPM is shown to have very low cytotoxic potency when added to extracellular culture medium and to be rapidly converted to other hydrophilic products including dichloro-isophosphoramide mustard (IPM). Only traces of Br-IPM or IPM were detected in the extracellular medium when generated by cellular metabolism of TH-302. We identify, in NfsA-expressing cells, the hydroxylamine metabolite of TH-302, and downstream products resulting from rearrangement or hydration of the imidazole ring, and demonstrate that formation of these candidate bystander effect mediators is suppressed by hypoxia. This characterisation of the cellular pharmacology of TH-302 implies that bystander effects from hypoxic activation of TH-302 are unlikely to contribute to its anticancer activity.


Asunto(s)
Antineoplásicos/farmacología , Efecto Espectador , Nitroimidazoles/farmacología , Mostazas de Fosforamida/farmacología , Proteínas de Escherichia coli , Células HCT116 , Humanos , Estructura Molecular , NADPH-Ferrihemoproteína Reductasa , Nitroimidazoles/química , Nitrorreductasas , Mostazas de Fosforamida/química , Profármacos/farmacología
16.
Eur J Med Chem ; 127: 442-458, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-28092860

RESUMEN

A series of novel compounds with phosphoramide mustard functionality incorporated into the quinazoline scaffold of EGFR/HER2 inhibitors were designed and synthesized as multi-target-directed ligands against tumor cells. In vitro assays showed that tumor cell lines with high HER2 level were more sensitive to the compounds than tumor cells with low HER2 level. Compound 10d (EMB-3) was one of the most potent inhibitors with IC50 of 7.4 nM and 82 nM against EGFR and HER2, respectively. The mechanism studies were also supported by the effect of 10d-induced DNA damage in MDA-MB-468 cells. In vivo efficacy study showed that 10d could significantly inhibit H522 tumor xenograft model with a TGI of 68% at dose of 100 mg/kg (QDx28, p.o.) and no significant body weight loss was observed. MTD study indicated that compound 10d had no acute toxicity to mice at doses up to 900 mg/kg (single dose).


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Diseño de Fármacos , Mostazas de Fosforamida/síntesis química , Mostazas de Fosforamida/farmacología , Quinazolinas/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Técnicas de Química Sintética , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones , Mostazas de Fosforamida/química , Mostazas de Fosforamida/farmacocinética , Ratas , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Med Chem ; 49(14): 4333-43, 2006 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16821793

RESUMEN

In efforts to obtain anticancer prodrugs for antibody-directed or gene-directed enzyme prodrug therapy using E. coli nitroreductase, a series of nitrobenzylphosphoramide mustards were designed and synthesized incorporating a strategically placed nitro group in a position para to the benzylic carbon for reductive activation. All analogues were good substrates of E. coli nitroreductase with half-lives between 2.9 and 11.9 min at pH 7.0 and 37 degrees C. Isomers of the 4-nitrophenylcyclophosphamide analogues 3 and 5 with a benzylic oxygen para to the nitro group showed potent selective cytotoxicity in nitroreductase (NTR) expressing cells, while analogues 4 and 6 with a benzylic nitrogen para to the nitro group showed little selective cytotoxicity despite their good substrate activity. These results suggest that good substrate activity and the benzylic oxygen are both required for reductive activation of 4-nitrophenylcyclophosphamide analogues by E. coli nitroreductase. Isomers of analogue 3 showed 23,000-29,000x selective cytotoxicity toward NTR-expressing V79 cells with an IC(50) as low as 27 nM. They are about as active as and 3-4x more selective than 5-aziridinyl-2,4-dinitrobenzamide (CB1954). The acyclic 4-nitrobenzylphosphoramide mustard ((+/-)-7) was found to be the most active and most selective compound for activation by NTR with 170,000x selective cytotoxicity toward NTR-expressing V79 cells and an IC(50) of 0.4 nM. Compound (+/-)-7also exhibited good bystander effect compared to 5-aziridinyl-2,4-dinitrobenzamide. The low IC(50), high selectivity, and good bystander effects of nitrobenzylphosphoramide mustards in NTR-expressing cells suggest that they could be used in combination with E. coli nitroreductase in enzyme prodrug therapy.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas de Escherichia coli/metabolismo , Nitrorreductasas/metabolismo , Mostazas de Fosforamida/síntesis química , Profármacos/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Cricetinae , Cricetulus , Diseño de Fármacos , Activación Enzimática , Proteínas de Escherichia coli/genética , Humanos , Nitrorreductasas/genética , Mostazas de Fosforamida/química , Mostazas de Fosforamida/farmacología , Profármacos/química , Profármacos/farmacología , Estereoisomerismo , Relación Estructura-Actividad , Transfección
18.
Artículo en Inglés | MEDLINE | ID: mdl-16455312

RESUMEN

Glufosfamide is a new, potential chemotherapeutic agent currently under investigation. Stability of glufosfamide was investigated in sodium phosphate buffers with different pH and temperature and in biological samples. Glufosfamide and isophosphamide mustard were quantified simultaneously using a liquid chromatography-ion trap mass spectrometric method; precision and accuracy were within 15% for each analyte. Glufosfamide was stable in neutral buffers, but decomposed to form isophosphoramide mustard under acidic and basic conditions, which was pH- and temperature-dependent. The stability of glufosfamide varied in different biological samples. Results indicated that glufosfamide was unstable in some biological samples, such as the small intestine, smooth muscles, pancreas and urine, especially in the small intestine homogenate, with a half-life of 1.1 h. But the pH (<8) and beta-glucosidase of the tissue homogenate was found to have negligible contribution to the degradation of glufosfamide. The enzymatic inhibition experiment with the specific inhibitor, saccharo-1,4-lactone, demonstrated that it was glucuronidase that resulted in the degradation of glufosfamide in small intestine homogenate. Methanol was recommended to be used to homogenize the tissue in an ice water bath, and the container for urine collection should also be maintained in an ice water bath, and all the biological samples collected should be preserved in frozen condition until analysis.


Asunto(s)
Antineoplásicos Alquilantes/química , Estabilidad de Medicamentos , Fosfatos/química , Mostazas de Fosforamida/química , Animales , Antineoplásicos Alquilantes/metabolismo , Tampones (Química) , Cromatografía Liquida , Glucosa/análogos & derivados , Ifosfamida/análogos & derivados , Espectrometría de Masas , Mostazas de Fosforamida/metabolismo , Ratas , Ratas Wistar
19.
Curr Pharm Des ; 5(8): 645-63, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10469896

RESUMEN

The nitrogen mustard based DNA alkylating agents were the first effective anticancer agents and remain important drugs against many forms of cancer. More than fifty years of research on the nitrogen mustards has yielded a broad range of therapeutically useful compounds and a detailed knowledge of the biochemical mechanism of these drugs. Nevertheless, there is much ongoing research on the phosphosphoramidic and other nitrogen mustards to increase their potency and reduce their toxic and mutagenic side effects. To understand the existing nitrogen mustards, and to design the next generation of these drugs, more knowledge is needed about the effects of chemical modifications on their activation and selectivity. Because of the existing knowledge of these drugs, atomic-level chemical modeling can play an important role in the understanding of the phosphoramidic mustard compounds; however, it has not proved straight forward to directly relate the activity of these mustards with simple chemical properties such as bond lengths or atomic charges. Instead, quantum chemical simulations will be required to simulate the activation and alkylation reactions of these compounds, which will require the newest generation of quantum chemical and solvent modeling methods. Additionally, molecular dynamics simulations of the adducted DNA can provide data on the factors favoring crosslinking and its structural consequences. This review summarizes the extensive literature on the metabolism, activation, and action of the phosphoramidic mustards, with an emphasis on the roles that chemical modeling has and will play in the development of this important class of drugs.


Asunto(s)
Antineoplásicos Alquilantes/química , Modelos Químicos , Mostazas de Fosforamida/química , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Simulación por Computador , Mostazas de Fosforamida/metabolismo , Mostazas de Fosforamida/farmacocinética , Mostazas de Fosforamida/farmacología
20.
J Med Chem ; 41(4): 515-29, 1998 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-9484502

RESUMEN

NMR (1H and 31P) and HPLC techniques were used to study the partitioning of phosphoramide mustard (PM) and its aziridinium ions among alkylation and P-N bond hydrolysis reactions as a function of the concentration and strength of added nucleophiles at 37 degrees C and pH 7.4. With water as the nucleophile, bisalkylation accounted for only 10-13% of the product distribution given by PM. The remainder of the products resulted from P-N bond hydrolysis reactions. With 50 mM thiosulfate or 55-110 mM glutathione (GSH), bisalkylation by a strong nucleophile increased to 55-76%. The rest of the PM was lost to either HOH alkylation or P-N bond hydrolysis reactions. Strong experimental and theoretical evidence was obtained to support the hypothesis that the P-N bond scission observed at neutral pH does not occur in the parent PM to produce nornitrogen mustard; rather it is an aziridinium ion derived from PM which undergoes P-N bond hydrolysis to give chloroethylaziridine. In every buffer studied (bis-Tris, lutidine, triethanolamine, and Tris), the decomposition of PM (with and without GSH) gave rise to 31P NMR signals which could not be attributed to products of HOH or GSH alkylation or P-N bond hydrolysis. The intensities of these unidentified signals were dependent on the concentration of buffer.


Asunto(s)
Aziridinas/química , Mostazas de Fosforamida/química , Alquilación , Cromatografía Líquida de Alta Presión , Hidrógeno , Hidrólisis , Cinética , Modelos Químicos , Resonancia Magnética Nuclear Biomolecular , Fósforo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda