Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Toxicol Appl Pharmacol ; 278(3): 249-58, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24823293

RESUMEN

DNA damage has been shown to induce autophagy, but the role of autophagy in the DNA damage response and cell fate is not fully understood. BO-1012, a bifunctional alkylating derivative of 3a-aza-cyclopenta[a]indene, is a potent DNA interstrand cross-linking agent with anticancer activity. In this study, BO-1012 was found to reduce DNA synthesis, inhibit S phase progression, and induce phosphorylation of histone H2AX on serine 139 (γH2AX) exclusively in S phase cells. Both CHK1 and CHK2 were phosphorylated in response to BO-1012 treatment, but only depletion of CHK1, but not CHK2, impaired BO-1012-induced S phase arrest and facilitated the entry of γH2AX-positive cells into G2 phase. CHK1 depletion also significantly enhanced BO-1012-induced cell death and apoptosis. These results indicate that BO-1012-induced S phase arrest is a CHK1-dependent pro-survival response. BO-1012 also resulted in marked induction of acidic vesicular organelle (AVO) formation and microtubule-associated protein 1 light chain 3 (LC3) processing and redistribution, features characteristic of autophagy. Depletion of ATG7 or co-treatment of cells with BO-1012 and either 3-methyladenine or bafilomycin A1, two inhibitors of autophagy, not only reduced CHK1 phosphorylation and disrupted S phase arrest, but also increased cleavage of caspase-9 and PARP, and cell death. These results suggest that cells initiate S phase arrest and autophagy as pro-survival responses to BO-1012-induced DNA damage, and that suppression of autophagy enhances BO-1012-induced apoptosis via disruption of CHK1-dependent S phase arrest.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinoma/tratamiento farmacológico , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Proteínas Quinasas/metabolismo , Antineoplásicos Alquilantes/agonistas , Proteína 7 Relacionada con la Autofagia , Carbamatos/agonistas , Carbamatos/farmacología , Carcinoma/enzimología , Carcinoma/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Fosfatidilinositol 3-Quinasas Clase III/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Femenino , Silenciador del Gen , Células HeLa , Compuestos Heterocíclicos con 3 Anillos/agonistas , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Indenos/agonistas , Indenos/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas/química , Proteínas Quinasas/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Fase S/efectos de los fármacos , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/metabolismo , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores , ATPasas de Translocación de Protón Vacuolares/metabolismo
2.
Int J Cancer ; 130(6): 1440-50, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21500194

RESUMEN

Oral cancer is the fourth-most common cause of death in males and overall the sixth-most common cause of cancer death in Taiwan. Surgery, radiotherapy and chemotherapy combined with other therapies are the most common treatments for oral cavity cancer. Although cisplatin, 5-fluorouracil and docetaxel are commonly used clinically, there is no drug specific for oral cavity cancer. Here, we demonstrated that derivatives of 3a-aza-cyclopenta[a]indene, a class of newly synthesized alkylating agents, may be drugs more specific for oral cancer based on its potent in vitro cytotoxicity to oral cancer cells and on in vivo xenografts. Among them, BO-1090, bis(hydroxymethyl)-3a-aza-cyclopenta[a]indene derivative, targeted DNA for its cytotoxic effects as shown by inhibition of DNA synthesis (bromodeoxyuridine-based DNA synthesis assay), induction of DNA crosslinking (alkaline gel shift assay), and induction of DNA single-stranded breaks (Comet assay) and double-stranded breaks (γ-H2AX focus formation). Following DNA damage, BO-1090 induced G1/S-phase arrest and apoptosis in oral cancer cell lines. The therapeutic potential of BO-1090 was tested in mice that received a xenograft of oral cavity cancer cell lines (SAS or Cal 27 cells). Intravenous injection of BO-1090 significantly suppressed tumor growth in comparison to control mice. BO-1090 also significantly reduced the tumor burden in orthotopic mouse models using SAS cells. There was no significant adverse effect of BO-1090 treatment with this dosage based on whole blood count, biochemical enzyme profiles in plasma and histopathology of various organs in mouse. Taken together, our current results demonstrate that B0-1090 may have potential as a treatment for oral cavity cancer.


Asunto(s)
Alquilantes/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , ADN/efectos de los fármacos , Neoplasias de la Boca/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Células CHO , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Cricetinae , Daño del ADN , Fibroblastos/efectos de los fármacos , Fase G1/efectos de los fármacos , Humanos , Células KB , Ratones , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Fase S/efectos de los fármacos
3.
Int J Mol Sci ; 13(3): 3277-3290, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22489152

RESUMEN

Although surgery or the combination of chemotherapy and radiation are reported to improve the quality of life and reduce symptoms in patients with oral cancer, the prognosis of oral cancer remains generally poor. DNA alkylating agents, such as N-mustard, play an important role in cancer drug development. BO-1051 is a new 9-anilinoacridine N-mustard-derivative anti-cancer drug that can effectively target a variety of cancer cell lines and inhibit tumorigenesis in vivo. However, the underlying mechanism of BO-1051-mediated tumor suppression remains undetermined. In the present study, BO-1051 suppressed cell viability with a low IC(50) in oral cancer cells, but not in normal gingival fibroblasts. Cell cycle analysis revealed that the tumor suppression by BO-1051 was accompanied by cell cycle arrest and downregulation of stemness genes. The enhanced conversion of LC3-I to LC3-II and the formation of acidic vesicular organelles indicated that BO-1501 induced autophagy. The expression of checkpoint kinases was upregulated as demonstrated with Western blot analysis, showing that BO-1051 could induce DNA damage and participate in DNA repair mechanisms. Furthermore, BO-1051 treatment alone exhibited a moderate tumor suppressive effect against xenograft tumor growth in immunocompromised mice. Importantly, the combination of BO-1051 and radiation led to a potent inhibition on xenograft tumorigenesis. Collectively, our findings demonstrated that BO-1051 exhibited a cytotoxic effect via cell cycle arrest and the induction of autophagy. Thus, the combination of BO-1051 and radiotherapy may be a feasible therapeutic strategy against oral cancer in the future.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias de la Boca/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/farmacología , Animales , Antineoplásicos Alquilantes/administración & dosificación , Autofagia/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2/metabolismo , Daño del ADN , Relación Dosis-Respuesta a Droga , Femenino , Encía/citología , Encía/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/metabolismo , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Compuestos de Mostaza Nitrogenada/administración & dosificación , Fosforilación , Proteínas Quinasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Bioorg Med Chem ; 19(1): 275-86, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21144756

RESUMEN

A series of linear pyrrolo[1,2-b]isoquinoline derivatives was synthesized for antitumor evaluation. The preliminary antitumor studies reveal that both bis(hydroxymethyl) and their bis(alkylcarbamate) derivatives show significant antitumor activity in inhibiting various human tumor cell growth in vitro. 1,2-Bis(hydroxymethyl)-3-methyl-5,10-dihydropyrrolo[1,2-b]isoquinoline (20a) was selected for antitumor studies in animal models. The results show that this agent can induce complete tumor remission or significant suppression in nude mice bearing human breast (MX-1) xenograft and ovarian (SK-OV-3) xenografts, respectively. Alkaline agarose gel shifting assay showed that 20a is able to cross-link with DNA. Studies on the cell cycle inhibition revealed that this agent induces cell arrest at G2/M phase. The results warrant further antitumor investigation against other human tumor growth in animal models.


Asunto(s)
Antineoplásicos Alquilantes/síntesis química , Antineoplásicos Alquilantes/farmacología , Isoquinolinas/síntesis química , Isoquinolinas/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Bioorg Med Chem ; 19(6): 1987-98, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21356592

RESUMEN

A series of N-mustard-quinazoline conjugates was synthesized and subjected to antitumor studies. The N-mustard pharmacophore was attached at the C-6 of the 4-anilinoquinazolines via a urea linker. To study the structure-activity relationships of these conjugates, various substituents were introduced to the C-4 anilino moiety. The preliminary antitumor studies revealed that these agents exhibited significant antitumor activity in inhibiting various human tumor cell growths in vitro. Compounds 21b, 21g, and 21h were selected for further antitumor activity evaluation against human breast carcinoma MX-1 and prostate PC-3 xenograft in animal model. These agents showed 54-75% tumor suppression with low toxicity (5-7% body-weight changes). We also demonstrate that the newly synthesized compounds are able to induce DNA cross-linking through alkaline agarose gel shift assay and inhibited cell cycle arrest at G2/M phase.


Asunto(s)
Antineoplásicos/síntesis química , Quinazolinas/química , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Ratones , Ratones Desnudos , Planta de la Mostaza/química , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad , Urea/química , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Bioorg Med Chem ; 19(1): 471-85, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21106377

RESUMEN

A series of novel water-soluble N-mustard-benzene conjugates bearing a urea linker were synthesized. The benzene moiety contains various hydrophilic side chains are linked to the meta- or para-position of the urea linker via a carboxamide or an ether linkage. The preliminary antitumor studies revealed that these agents exhibited potent cytotoxicity in vitro and therapeutic efficacy against human tumor xenografts in vivo. Remarkably, complete tumor remission in nude mice bearing human breast carcinoma MX-1 xenograft and significant suppression against prostate adenocarcinoma PC3 xenograft were achieved by treating with compound 9aa' at the maximum tolerable dose with relatively low toxicity. We also demonstrate that the newly synthesized compounds are able to induce DNA cross-linking through alkaline agarose gel shift assay. A pharmacokinetic profile of the representative 9aa' in rats was also investigated. The current studies suggest that this agent is a promising candidate for preclinical studies.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Diseño de Fármacos , Compuestos de Mostaza/síntesis química , Compuestos de Mostaza/farmacología , Animales , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Límite de Detección , Masculino , Ratones , Ratones Desnudos , Compuestos de Mostaza/química , Ratas , Ratas Sprague-Dawley , Solubilidad , Agua , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Med Chem ; 64(17): 12469-12486, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34459195

RESUMEN

Designing hybrid molecules with dual functions is one approach to improve the therapeutic efficacy of combination treatment. We have previously conjugated phthalazine and bis(hydroxymethyl)pyrrole pharmacophores to form hybrids bearing antiangiogenesis and DNA interstrand cross-linking activities. To improve the bioavailability, we adopted a benzology approach to design and synthesize a new series of 1,2-bis(hydroxymethyl)benzo[g]pyrrolo[2,1-a]phthalazines. These new hybrids retained the dual functions and could be formulated into vehicles for intravenous and oral administration. Among them, we demonstrated that compound 19a with dimethylamine at the C6 position markedly suppressed the tumor growth of human small cell lung cancer cell line H526, squamous lung cancer cell line H520, and renal cancer cell line 786-O in nude mice, implying that compound 19a is a broad-spectrum anticancer agent. Our results implicated that the conjugation of antiangiogenic and DNA cross-linking is likely to be a helpful approach to improving the efficacy of combination therapy.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neovascularización Patológica/prevención & control , Ftalazinas/química , Ftalazinas/farmacología , Animales , Línea Celular Tumoral , Supervivencia Celular , Diseño de Fármacos , Humanos , Neoplasias Pulmonares , Ratones , Ratones Desnudos , Neoplasias de Células Escamosas , Carcinoma Pulmonar de Células Pequeñas , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Int J Cancer ; 126(4): 1017-28, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19697325

RESUMEN

Glyfoline exhibits cytotoxic activity in vitro and antitumor activity in mice bearing murine or human solid tumors, but the underlying mechanisms are unknown. In our study, we found that glyfoline inhibited cell growth and induced accumulation of mitotic cells in human cancer cell lines. Glyfoline induced the appearance of spindle abnormalities, chromosome mis-segregation, multipolar cell division and multiple nuclei, all of which are indicative of mitotic catastrophe. However, glyfoline did not bind to DNA and did not inhibit or stabilize tubulin polymerization, but slightly increased the resistance of mitotic spindles to nocodazole-induced disassembly. In addition, microtubule aster formation was significantly enhanced in the extract prepared from glyfoline-arrested mitotic cells compared to that from synchronized mitotic cells. When Eg5, a mitotic kinesin that plays an essential role in establishing mitotic spindle bipolarity, was inhibited using S-trityl-cysteine in glyfoline-treated cells, formation of spindle multipolarity, multipolar cell division, and multinuclei was significantly reduced. After glyfoline-mediated arrest of cells at mitosis, considerable poly(ADP-ribose) polymerase degradation was induced and the number of annexin V-positive cells significantly increased, indicating that glyfoline ultimately induces apoptosis. Small interfering RNA-mediated silencing of the spindle checkpoint proteins BUBR1 and MAD2 markedly reduced induction of mitotic cell accumulation, but did not affect glyfoline-induced mitotic catastrophe and apoptosis. Thus, glyfoline induces mitotic catastrophe probably by enhancing microtubule aster formation and subsequent apoptosis in cancer cells independently of spindle checkpoint function.


Asunto(s)
Acridinas/farmacología , Apoptosis/efectos de los fármacos , Mitosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Antineoplásicos/uso terapéutico , División Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Células HeLa/efectos de los fármacos , Células HeLa/patología , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Microtúbulos/efectos de los fármacos , Neoplasias/patología , Neoplasias de la Próstata/patología , Tubulina (Proteína)/efectos de los fármacos , Tubulina (Proteína)/metabolismo
9.
Bioorg Med Chem ; 18(6): 2285-2299, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20181487

RESUMEN

A series of N-mustard-quinoline conjugates bearing a urea or hydrazinecarboxamide linker was synthesized for antitumor evaluation. The in vitro cytotoxicity studies revealed that compounds with hydrazinecarboxamide linkers were generally more cytotoxic than the corresponding urea counterparts in inhibiting human lymphoblastic leukemia and various solid tumor cell growths in culture. The therapeutic efficacy against human tumor xenografts in animal model was studied. It was shown that complete tumor remission in nude mice bearing human breast carcinoma MX-1 xenograft by 17a, i and 18c, d was achieved. In the present study, it was revealed that both linkers are able to lower the chemically reactive N-mustard pharmacophore and thus the newly synthesized conjugates possess a long half-life in rat plasma. Moreover, the new N-mustard derivatives are able to induce DNA cross-linking either by modified comet assay or by alkaline agarose gel shift assay.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , ADN/química , ADN/efectos de los fármacos , Hidrazinas/química , Neoplasias Experimentales/tratamiento farmacológico , Quinolinas/farmacología , Urea/química , Animales , Antineoplásicos Alquilantes/síntesis química , Antineoplásicos Alquilantes/química , Proliferación Celular/efectos de los fármacos , ADN/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Ratones , Ratones Desnudos , Estructura Molecular , Trasplante de Neoplasias , Quinolinas/síntesis química , Quinolinas/química , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
10.
Eur J Med Chem ; 202: 112516, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32622270

RESUMEN

A series of 1,2-bis(hydroxymethyl)pyrrolo[1,2-f]phenanthridine derivatives and their alkyl (ethyl and isopropyl) carbamates and 12,13-bis(hydroxymethyl)-9,14-dihydro-dibenzo[f,h]pyrrolo[1,2-b]isoquinoline derivatives were synthesized for antiproliferative evaluation. The preliminary antitumour studies revealed that these two types of bis(hydroxymethyl) derivatives showed significant antitumour activities and were able to inhibit the growth of various human tumour cell lines in vitro. Several of the derivatives were demonstrated to cause DNA interstrand cross-links by an alkaline agarose gel shifting assay. These conjugates were cytotoxic to a variety of cancer cell lines by inducing DNA damage, delaying cell cycle progression in the G2/M phase and triggering apoptosis. Compound 21a, dissolved in a vehicle suitable for intravenous administration, was selected for antitumour studies in animal models. We demonstrated that at a dose that did not cause body weight loss in mice, compound 21a could significantly suppress the growth of tumour xenografts of human lung cancer H460 and colorectal cancer HCT-116 cells in nude mice. Our present results confirm the antitumour activities of these conjugates.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Isoquinolinas/farmacología , Fenantridinas/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inyecciones Intravenosas , Isoquinolinas/administración & dosificación , Isoquinolinas/química , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Fenantridinas/administración & dosificación , Fenantridinas/química , Relación Estructura-Actividad
11.
Bioorg Med Chem ; 17(3): 1264-75, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19124250

RESUMEN

A series of phenyl N-mustard-9-anilinoacridine conjugates via a carbamate or carbonate linker was synthesized for antitumor evaluation. The carbamate or carbonate linker is able to lower the reactivity of the phenyl N-mustard pharmacophore and thus, these conjugates are rather chemically stable. The in vitro studies revealed that these derivatives possessed significant cytotoxicity with IC(50) in sub-micromolar range in inhibiting human lymphoblastic leukemia (CCRF-CEM), breast carcinoma (MX-1), colon carcinoma (HCT-116) and human non-small cell lung cancer (H1299) cell growth in vitro. Compounds 10a, 10b, 10e, 10i, and 15a were selected for evaluating their antitumor activity in nude mice bearing MX-1 and HCT-116 xenografts. Remarkably, total tumor remission was achieved by these agents with only one cycle of treatment. Interestingly, no tumor relapse was found in mice treated with 10a over 129 days. This agent is capable of inducing DNA interstrand cross-linking in human non-small lung cancer H1299 cells in a dose dependent manner by modified comet assay and has a long half-life in rat plasma.


Asunto(s)
Acridinas/química , Acridinas/farmacología , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacología , Carbamatos/química , Carbonatos/química , Acridinas/uso terapéutico , Animales , Antineoplásicos Alquilantes/uso terapéutico , Línea Celular Tumoral , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Semivida , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Bioorg Med Chem ; 17(15): 5614-26, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19576785

RESUMEN

A series of bifunctional DNA interstrand cross-linking agents, bis(hydroxymethyl)- and bis(carbamates)-8H-3a-azacyclopenta[a]indene-1-yl derivatives were synthesized for antitumor evaluation. The preliminary antitumor studies revealed that these agents exhibited potent cytotoxicity in vitro and antitumor therapeutic efficacy against human tumor xenografts in vivo. Furthermore, these derivatives have little or no cross-resistance to either Taxol or Vinblastine. Remarkably, complete tumor remission in nude mice bearing human breast carcinoma MX-1 xenograft by 13a,b and 14g,h and significant suppression against prostate adenocarcinoma PC3 xenograft by 13b were achieved at the maximum tolerable dose with relatively low toxicity. In addition, these agents induce DNA interstrand cross-linking and substantial G2/M phase arrest in human non-small lung carcinoma H1299 cells. The current studies suggested that these agents are promising candidates for preclinical studies.


Asunto(s)
Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/uso terapéutico , Reactivos de Enlaces Cruzados/farmacología , ADN/metabolismo , Indenos/química , Indenos/farmacología , Animales , Antineoplásicos Alquilantes/síntesis química , Antineoplásicos Alquilantes/farmacología , Compuestos Aza/síntesis química , Compuestos Aza/química , Compuestos Aza/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Reactivos de Enlaces Cruzados/síntesis química , Reactivos de Enlaces Cruzados/química , ADN/química , Femenino , Humanos , Indenos/síntesis química , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Inducción de Remisión , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Med Chem ; 62(5): 2404-2418, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30776229

RESUMEN

Hybrid molecules are composed of two pharmacophores with different biological activities. Here, we conjugated phthalazine moieties (antiangiogenetic pharmacophore) and bis(hydroxymethyl)pyrrole moieties (DNA cross-linking agent) to form a series of bis(hydroxymethyl)pyrrolo[2,1- a]phthalazine hybrids. These conjugates were cytotoxic to a variety of cancer cell lines by inducing DNA damage, arresting cell cycle progression at the G2/M phase, triggering apoptosis, and inhibiting vascular endothelial growth factor receptor 2 (VEGFR-2) in endothelial cells. Among them, compound 29d encapsulated in a liposomal formulation (e.g., 29dL) significantly suppressed the growth of small-cell lung cancer cell (H526) xenografts in mice. Based on immunohistochemical staining, the tumor xenografts in mice treated with 29dL showed time-dependent decreases in the intensity of CD31, a marker of blood vessels, whereas the intensity of γ-H2AX, a marker of DNA damage, increased. The present data revealed that the conjugation of antiangiogenic and DNA-damaging agents can generate potential hybrid agents for cancer treatment.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Daño del ADN/efectos de los fármacos , Ftalazinas/química , Ftalazinas/farmacología , Inhibidores de la Angiogénesis/síntesis química , Animales , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Ratones , Neovascularización Patológica/prevención & control , Fosforilación , Ftalazinas/síntesis química , Relación Estructura-Actividad , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Bioorg Med Chem ; 16(10): 5413-23, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18450456

RESUMEN

To improve the chemical stability and therapeutic efficacy of N-mustard, a series of phenyl N-mustard linked to DNA-affinic 9-anilinoacridines and acridine via a urea linker were synthesized and evaluated for antitumor studies. The new N-mustard derivatives were prepared by the reaction of 4-bis(2-chloroethyl)aminophenyl isocyanate with a variety of 9-anilinoacridines or 9-aminoacridine. The antitumor studies revealed that these agents exhibited potent cytotoxicity in vitro without cross-resistance to taxol or vinblastine and showed potent antitumor therapeutic efficacy in nude mice against human tumor xenografts. It also showed that 24d was capable of inducing marked dose-dependent levels of DNA cross-linking by comet assay and has long half-life in rat plasma.


Asunto(s)
Amsacrina/análogos & derivados , Mostaza de Anilina/síntesis química , Mostaza de Anilina/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Urea/química , Acridinas/química , Amsacrina/química , Mostaza de Anilina/análogos & derivados , Animales , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Desnudos , Estructura Molecular , Ratas , Estereoisomerismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Neoplasia ; 20(2): 119-130, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29247884

RESUMEN

Efficacy and safety are fundamental prerequisites for anticancer drug development. In the present study, we explored the anti-colorectal cancer (CRC) activity of SL-1, a DNA-directed N-mustard-quinoline conjugate. The N-mustard moiety in SL-1 induced DNA strand breaks, interstrand cross-links (ICLs), G2/M arrest, and apoptosis, whereas its quinoline moiety preferentially directed SL-1 to target the selective guanine sequence 5'-G-G/C-N-G-C/T-3'. Notably, SL-1 was highly cytotoxic to various CRC cell lines. Experiments using xenograft models revealed that SL-1 was more potent than 5-fluorouracil (5-FU) and oxaliplatin for suppressing the growth of RKO and RKO-E6 (oxaliplatin-resistant subline) cells as well as metastatic SW620 cells. In addition, SL-1 combined with 5-FU was more effective than oxaliplatin and 5-FU for suppressing RKO or SW620 cell growth in mice. Significantly, compared with cisplatin, oxaliplatin, or 5-FU, SL-1 alone or in combination with 5-FU did not cause obvious kidney or liver toxicity in ICR mice. In summary, SL-1, a DNA-directed alkylating agent, is established as an anti-CRC agent with high efficacy and low toxicity and thus warrants further development for the treatment of CRC patients.


Asunto(s)
Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacología , Antineoplásicos/farmacología , Neoplasias Colorrectales/patología , ADN/química , Compuestos de Mostaza/química , Compuestos de Mostaza/farmacología , Quinolinas/química , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , ADN/genética , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Quinolinas/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Oncotarget ; 8(28): 45072-45087, 2017 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-28178691

RESUMEN

Recent studies have demonstrated that P-glycoprotein (P-gp) expression impairs DNA interstrand cross-linking agent-induced DNA repair efficiency in multidrug-resistant (MDR) cells. To date, the detailed molecular mechanisms underlying how P-gp interferes with Src activation and subsequent DNA repair activity remain unclear. In this study, we determined that the C-terminal Src kinase-binding protein (Cbp) signaling pathway involved in the negative control of Src activation is enhanced in MDR cells. We also demonstrated that cells that ectopically express P-gp exhibit reduced activation of DNA damage response regulators, such as ATM, Chk2, Braca1 and Nbs1 and hence attenuated DNA double-strand break repair capacity and become more susceptible than vector control cells to DNA interstrand cross-linking (ICL) agents. Moreover, we demonstrated that P-gp can not only interact with Cbp and Src but also enhance the formation of inhibitory C-terminal Src kinase (Csk)-Cbp complexes that reduce phosphorylation of the Src activation residue Y416 and increase phosphorylation of the Src negative regulatory residue Y527. Notably, suppression of Cbp expression in MDR cells restores cisplatin-induced Src activation, improves DNA repair capacity, and increases resistance to ICL agents. Ectopic expression of Cbp attenuates cisplatin-induced Src activation and increases the susceptibility of cells to ICL agents. Together, the current results indicate that P-gp inhibits DNA repair activity by modulating Src activation via Cbp-Csk-Src cascade. These results suggest that DNA ICL agents are likely to have therapeutic potential against MDR cells with P-gp-overexpression.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Reparación del ADN , Familia-src Quinasas/metabolismo , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Cisplatino/farmacocinética , Cisplatino/farmacología , Resistencia a Antineoplásicos , Humanos , Células KB , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Anticancer Agents Med Chem ; 17(13): 1741-1755, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28530540

RESUMEN

BACKGROUND: Bendamustine, an N-mustard-benzoimidazole hybrid conjugate, was recently approved for the treatment of chronic lymphocytic leukemia. However, the short half-life of bendamustine may limit its clinical applications. OBJECTIVE: The purpose of this study is to design and synthesize compounds with a more favorable pharmacokinetic profile. METHODS: We synthesized a series of hybrid molecules comprising a phenyl N-mustard moiety and benzothiazole or benzimidazole scaffold linked via a urea linker and evaluated their antitumor activity and plasma stability. RESULTS: We revealed that these agents exhibited significant cytotoxicity against a panel of human lymphoblastic leukemia and human solid tumor cells in culture. Human lymphoblastic leukemia CCRM-CEM cells were the most sensitive to the tested compounds. In general, the new hybrids were as potent as cisplatin, but significantly more cytotoxic than bendamustine. Phenyl N-mustard-benzothiazole compound 27d and phenyl N-mustardbenzimidaloe compound 32b possessed significant cytotoxicity and led to apoptotic death in the treated tumor cells. These two agents were able to induce DNA interstrand cross-linking and arrested cell cycle progression at the G2/M phase. Furthermore, we showed that these new hybrids were more chemically stable than bendamustine in rat plasma. CONCLUSION: Our results suggest that conjugation of phenyl N-mustard pharmacophore at C6 of benzimidazole or at C8 of the benzothiazole ring via a urea linker is likely an approach to increase the chemical stability and bioavailability. Highlights ⇒ Series of benzimidazoles and benzothiazoles linked to N-mustard were synthesized. ⇒ The newly synthesized derivatives induced DNA interstrand cross-links. ⇒ These derivatives induced cell cycle arrest in the G2/M phase and triggered apoptosis in H460 cells. ⇒ The new compounds are more cytotoxic than bendamustine. ⇒ The new compounds were chemically more stable than bendamustine in rat plasma.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Bencimidazoles/síntesis química , Bencimidazoles/farmacología , Benzotiazoles/síntesis química , Benzotiazoles/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Bencimidazoles/química , Benzotiazoles/química , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Humanos , Ratas
18.
Eur J Med Chem ; 127: 235-249, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-28064078

RESUMEN

A novel series of bis(hydroxymethyl)indolizino[8,7-b]indole hybrids composed of ß-carboline (topoisomerase I/II inhibition) and bis(hydroxymethyl)pyrrole (DNA cross-linking) are synthesized for antitumor evaluation. Of tumor cell lines tested, small cell lung cancer (SCLC) cell lines are the most sensitive to the newly synthesized compounds. These hybrids induce cell cycle arrest at the G2/M phase, trigger tumor cell apoptotic death, and display diverse mechanisms of action involving topoisomerase II (Topo II) inhibition and induction of DNA cross-linking. Intriguingly, the substituent at N11 (H or Me) plays a critical role in modulating Topo II inhibition and DNA cross-linking activities. N11-Me derivatives predispose to induce DNA crosslinks, whereas N11-H derivatives potently inhibit Topo II. Computational analysis implicates that N11-Me restrict the torsion angles of the two adjacent OH on pyrrole resulting in a favorable of DNA cross-linking. Among these hybrids, compound 17a with N11-H is more effective than cisplatin and etoposide, but as potent as irinotecan, against the growth of SCLC H526 cells in xenograft model.


Asunto(s)
ADN-Topoisomerasas de Tipo II/metabolismo , ADN/metabolismo , Diseño de Fármacos , Indoles/síntesis química , Indoles/farmacología , Neoplasias Pulmonares/patología , Carcinoma Pulmonar de Células Pequeñas/patología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Indoles/química , Indoles/metabolismo , Ratones , Inhibidores de Topoisomerasa I/síntesis química , Inhibidores de Topoisomerasa I/química , Inhibidores de Topoisomerasa I/metabolismo , Inhibidores de Topoisomerasa I/farmacología , Inhibidores de Topoisomerasa II/síntesis química , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/metabolismo , Inhibidores de Topoisomerasa II/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Med Chem ; 49(12): 3710-8, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16759114

RESUMEN

A series of 9-anilinoacridine and acridine derivatives bearing an alkylating N-mustard residue at C4 of the acridine chromophore were synthesized. The N-mustard pharmacophore was linked to the C4 of the acridine ring with an O-ethyl (O-C(2)), O-propyl (O-C(3)), or O-butyl (O-C(4)) spacer. It revealed that all newly synthesized compounds were very potent cytotoxic agents against human leukemia and various solid tumors in vitro. These agents did not exhibit cross-resistance against vinblastine-resistant (CCRF-CEM/VBL) or taxol-resistant (CCRF-CEM/taxol) cells. It also showed that these agents were DNA cross-linking agents rather than topoisomerase II inhibitors. Of these agents, compounds 27a and 27c were shown to have potent antitumor activity in nude mice bearing the human breast carcinoma MX-1 xenograft. The therapeutic efficacies of these two agents are comparable to that of taxol.


Asunto(s)
Acridinas/síntesis química , Aminoacridinas/síntesis química , Compuestos de Anilina/síntesis química , Antineoplásicos Alquilantes/síntesis química , Compuestos de Mostaza Nitrogenada/síntesis química , Acridinas/química , Acridinas/farmacología , Aminoacridinas/química , Aminoacridinas/farmacología , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Animales , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , ADN/química , ADN-Topoisomerasas de Tipo II/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Compuestos de Mostaza Nitrogenada/química , Compuestos de Mostaza Nitrogenada/farmacología , Relación Estructura-Actividad , Trasplante Heterólogo
20.
Oncotarget ; 7(25): 38078-38090, 2016 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-27191263

RESUMEN

Autophagy is a lysosomal degradative process that protects cancer cells from multiple types of stress. In this study, we synthesized a series of derivatives of 6-cinnamamido-quinoline-4-carboxamide (CiQ), and investigated their effects on the proliferation and autophagy of cancer cells in vitro. These derivatives effectively inhibited the proliferation of a broad spectrum of cancer cell lines. Further study revealed that CiQ derivatives may induce autophagy and result in disruption of autophagy propagation. Consequently, these derivatives triggered massive apoptosis, as evidenced by caspase-9 activation and PARP cleavage. Blockage of autophagy by depletion of autophagy related gene ATG5 or BECN1 considerably alleviated CiQ-induced cell death, indicating that autophagy may mediate CiQ-induced cell death. Furthermore, treatment with CiQ derivatives increased lysosome membrane permeability (LMP) and enhanced accumulation of ubiquitinated proteins, which collectively indicate impaired lysosome function. In addition, treatment of cells with CiQ derivatives activated extracellular signal-regulated kinase (ERK); abrogation of ERK activation, either by treating cells with U0126, an inhibitor of mitogen-activated protein/ERK kinase 1 (MEK1), or by ectopically overexpressing a dominant-negative MEK1, significantly reduced CiQ derivative-induced LMP, LC3 and p62 accumulation, and cytotoxicity. These results indicate that CiQ derivatives activate ERK and disrupt lysosome function, thereby altering autophagic flux and resulting in apoptotic cell death.


Asunto(s)
Autofagia/efectos de los fármacos , Quinolinas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Neoplasias/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA