Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Nanomedicine ; 19: 3805-3825, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38708177

RESUMEN

Telomere is a protective structure located at the end of chromosomes of eukaryotes, involved in maintaining the integrity and stability of the genome. Telomeres play an essential role in cancer progression; accordingly, targeting telomere dynamics emerges as an effective approach for the development of cancer therapeutics. Targeting telomere dynamics may work through multifaceted molecular mechanisms; those include the activation of anti-telomerase immune responses, shortening of telomere lengths, induction of telomere dysfunction and constitution of telomerase-responsive drug release systems. In this review, we summarize a wide variety of telomere dynamics-targeted agents in preclinical studies and clinical trials, and reveal their promising therapeutic potential in cancer therapy. As shown, telomere dynamics-active agents are effective as anti-cancer chemotherapeutics and immunotherapeutics. Notably, these agents may display efficacy against cancer stem cells, reducing cancer stem levels. Furthermore, these agents can be integrated with the capability of tumor-specific drug delivery by the constitution of related nanoparticles, antibody drug conjugates and HSA-based drugs.


Asunto(s)
Antineoplásicos , Neoplasias , Telomerasa , Telómero , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Telómero/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Telomerasa/antagonistas & inhibidores , Animales , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Inmunoterapia/métodos , Células Madre Neoplásicas/efectos de los fármacos
2.
J Cancer Res Ther ; 19(6): 1603-1609, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38156928

RESUMEN

OBJECTIVE: This study investigated the antitumor efficacy of programmed cell death protein-1 (PD-1) antibody and DBDx, a triple-drug combination of dipyridamole, bestatin, and dexamethasone, and their related immunomodulation. MATERIALS AND METHODS: Mouse melanoma B16, mouse Lewis lung carcinoma, and mouse breast carcinoma 4T1 were used for evaluating the in vivo therapeutic efficacy of DBDx, PD-1 antibody, and their combination. The peripheral blood and tumor tissues of 4T1 tumor-bearing mice were collected to analyze regulatory T cells and measured using flow cytometry. RESULTS: The combination of PD-1 antibody and DBDx enhanced the therapeutic efficacy against B16 melanoma. The suppression of tumor growth by PD-1 antibody and DBDx was more significant than that by anti-PD-1 monotherapy. The tumor growth inhibition rates of PD-1 antibody, DBDx, and their combination were 54.0%, 72.4%, and 83.1%, respectively, suggesting a synergistic effect as determined by the coefficient of drug interaction. No significant changes were found in the body weights in all the above groups, indicating that the treated mice tolerated the applied drug doses. Similarly, enhanced therapeutic efficacy of the PD-1 antibody and DBDx combination was observed in murine Lewis lung carcinoma and 4T1 breast cancer models. In 4T1 breast cancer-bearing mice, the immunotherapy-related changes in lymphocytes in peripheral blood and tumor microenvironment were evaluated with flow cytometry. Compared with anti-PD-1 monotherapy, peripheral blood and tumor-infiltrating lymphocytes were found a lower ratio of regulatory T cell (Treg) subset cells and a higher ratio of CD8+/Treg cells. CONCLUSIONS: The combination of PD-1 antibody and DBDx could achieve enhanced therapeutic antitumor efficacy than anti-PD-1 monotherapy, suggesting potential for using the triple-drug combination DBDx in cancer immunotherapy.


Asunto(s)
Carcinoma Pulmonar de Lewis , Linfocitos T Reguladores , Animales , Ratones , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Receptor de Muerte Celular Programada 1 , Línea Celular Tumoral , Linfocitos T CD8-positivos , Combinación de Medicamentos , Microambiente Tumoral
3.
Int J Biol Macromol ; 226: 1088-1099, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36435475

RESUMEN

OBJECTIVE: To prepare a recombinant EGFR-targeted fusion protein drug conjugate acting on telomere and telomerase; and evaluate its antitumor efficacy. METHODS: We prepared a recombinant fusion protein Fv-LDP-D3 which consists of the Fv fragment of an anti-EGFR monoclonal antibody (MAb), the apoprotein of lidamycin (LDP), and the third domain (D3) of human serum albumin (HSA); then generated the conjugate Fv-LDP-D3∼AE by integrating the active enediyne chomophore (AE) of lidamycin. Accordingly, in vitro and in vivo experiments were performed. RESULTS: As shown, Fv-LDP-D3 specifically bound to EGFR highly-expressing cancer cells and intensely entered K-Ras mutant cells via enhanced macropinocytosis. By in vivo imaging, Fv-LDP-D3 displayed intense accumulation and persistent retention in tumor-site. Furthermore, the conjugate Fv-LDP-D3∼AE displayed highly potent cytotoxicity to cancer cells with IC50 at 0.1 nM level. The conjugate induced telomere shortening and downregulation of telomerase and EGFR pathway related proteins. Fv-LDP-D3∼AE exhibited prominent antitumor efficacy against human colorectal cancer xenograft accompanying with significant increase of serum IFN-ß in athymic mice. CONCLUSION: The recombinant fusion protein conjugate that exhibits the capability of tumor-targeting drug delivery can induce telomere shortening and telomerase downregulation. The investigation may lay the foundation for the development of MAb-HSA domain-based fusion protein drug conjugates.


Asunto(s)
Inmunoconjugados , Telomerasa , Animales , Ratones , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Receptores ErbB/metabolismo , Regulación hacia Abajo , Acortamiento del Telómero , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Inmunoconjugados/farmacología , Telómero/metabolismo
4.
Biochem Pharmacol ; 201: 115057, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35489393

RESUMEN

KRAS mutation and NF-κB both play crucial role in pancreatic cancer; in addition, defensin, the peptide mediator in innate immunity, can inhibit NF-κB. Assuming a strategy that targets both NF-κB and concomitantly the mutated KRAS indirectly via intensive macropinocytosis, we designed and generated a recombinant protein DF2-HSA which consists of two molecules of human beta-defensin 2 (HBD2) and a moiety of human serum albumin (HSA). As shown, the recombinant protein DF2-HSA markedly down-regulated NF-κB in both KRAS mutant MIA PaCa-2 cells and wild type BxPC-3 cells. Determined by confocal microscopy, the uptake of DF2-HSA in MIA PaCa-2 cells was more intense than that in BxPC-3 cells. The uptake was blocked by the specific inhibitor EIPA, indicating that DF2-HSA internalized via macropinocytosis. DF2-HSA displayed more potent cytotoxicity to cancer cells than HBD2. DF2-HSA induced apoptosis in cancer cells. Notably, DF2-HSA inhibited tumor cell spheroid formation, an effect comparable to that of salinomycin. DF2-HSA inhibited tumor cell migration and invasion. As detected with scanning electron microscopy, DF2-HSA strongly depleted filopodia on cell surface; and salinomycin induced similar changes. By in vivo imaging, DF2-HSA displayed intense tumor-site accumulation and lasting retention for over 14 days; however, HBD2 showed much less tumor-site accumulation and a shorter retention time for only 24 h. DF2-HSA suppressed the growth of pancreatic cancer MIA PaCa-2 xenograft in athymic mice; and its combination with gemcitabine achieved higher antitumor efficacy. In summary, the recombinant defensin/HSA fusion protein that inhibits NF-κb associated with intensive macropinocytosis is highly effective against pancreatic cancer.


Asunto(s)
FN-kappa B , Neoplasias Pancreáticas , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , FN-kappa B/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Albúmina Sérica Humana/metabolismo , Albúmina Sérica Humana/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
5.
Drug Deliv ; 29(1): 1243-1256, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35416106

RESUMEN

The present study aimed to evaluate the anti-tumor efficacy of the epidermal growth factor receptor (EGFR)-targeting recombinant fusion protein Fv-LDP-D3 and its antibody-drug conjugate Fv-LDP-D3-AE against esophageal cancer. Fv-LDP-D3, consisting of the fragment variable (Fv) of an anti-EGFR antibody, the apoprotein of lidamycin (LDP), and the third domain of human serum albumin (D3), exhibited a high binding affinity for EGFR-overexpressing esophageal cancer cells, inhibited EGFR phosphorylation and down-regulated inosine monophosphate dehydrogenase type II (IMPDH2) expression. Fv-LDP-D3 was taken up by cancer cells through intensive macropinocytosis; it inhibited the proliferation and induced the apoptosis of esophageal cancer cells. In vivo imaging revealed that Fv-LDP-D3 displayed specific tumor-site accumulation and a long-lasting retention over a 26-day period. Furthermore, Fv-LDP-D3-AE, a pertinent antibody-drug conjugate prepared by integrating the enediyne chromophore of lidamycin into the Fv-LDP-D3 molecule, displayed highly potent cytotoxicity, inhibited migration and invasion, induced apoptosis and DNA damage, arrested cells at G2/M phase, and caused mitochondrial damage in esophageal cancer cells. More importantly, both of Fv-LDP-D3 and Fv-LDP-D3-AE markedly inhibited the growth of esophageal cancer xenografts in athymic mice at well tolerated doses. The present results indicate that Fv-LDP-D3, and Fv-LDP-D3-AE exert prominent antitumor efficacy associated with targeting EGFR, suggesting their potential as promising candidates for targeted therapy against esophageal cancer.


Asunto(s)
Neoplasias Esofágicas , Inmunoconjugados , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Enediinos/química , Enediinos/farmacología , Receptores ErbB/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Humanos , IMP Deshidrogenasa/genética , IMP Deshidrogenasa/metabolismo , IMP Deshidrogenasa/uso terapéutico , Inmunoconjugados/metabolismo , Inmunoconjugados/farmacología , Ratones , Ratones Desnudos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Int J Biol Macromol ; 187: 24-34, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34284054

RESUMEN

Human serum albumin (HSA)-based therapeutics have attracted tremendous attention in the development of anticancer agents. The versatile properties of HSA make HSA-based therapeutics possess improved pharmacokinetics, extended circulation half-life, enhanced efficacy, reduced toxicity, etc. Generally, the HSA-based therapeutics systems can be divided into four categories, i.e. HSA-drug nanoparticles, HSA-drug conjugates, HSA-binding prodrugs, and HSA-based recombinant fusion proteins: the latter mainly include antibody (domain)- and cytokine- fusion proteins. Advances in this area revealed the advantages of HSA-based systems in the development of tumor site-oriented therapeutics, partly referring to the enhanced penetration and retention (EPR) effect and the intensive macropinocytosis. Accordingly, a variety of technical platforms for the design and preparation of HSA-based therapeutics have been reported. Major strategies and directions for the drug development were discussed; those include (1) Tumor-site oriented drug delivery and enhanced drug retention, (2) Tumor-site prodrug release and activation, (3) Cancer cell bound intensive drug internalization, and (4) Tumor microenvironment (TME) directed immunomodulation. Notably, the multimodal HSA-based approach is promising for the development of tumor-oriented therapeutics for cancer therapy.


Asunto(s)
Antineoplásicos , Sistemas de Liberación de Medicamentos , Nanopartículas , Neoplasias/tratamiento farmacológico , Profármacos , Proteínas Recombinantes de Fusión , Albúmina Sérica Humana , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Humanos , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias/metabolismo , Neoplasias/patología , Profármacos/química , Profármacos/uso terapéutico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/uso terapéutico , Albúmina Sérica Humana/química , Albúmina Sérica Humana/genética , Albúmina Sérica Humana/uso terapéutico
7.
Oncol Rep ; 43(3): 851-863, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32020213

RESUMEN

Efficient enrichment and transmembrane transport of cytotoxic reagents are considered to be effective strategies to increase the efficiency and selectivity of antitumor drugs targeting solid tumors. In the present study, a recombinant protein ABD­LDP­Ec consisting of the albumin­binding domain (ABD), the apoprotein (LDP) of lidamycin (LDM) and an EGFR­targeting oligopeptide (Ec) was prepared by DNA recombination and bacterial fermentation, and was integrated with the enediyne chromophore (AE) of lidamycin to generate its enediyne­integrated analogue ABD­LDP­Ec­AE. ABD­LDP­Ec exhibited high binding capacity to both albumin and EGFR­positive pancreatic cancer cells, and was internalized into the cytoplasm through receptor­mediated endocytosis and albumin­driven macropinocytosis of K­ras mutant cells. In animal experiments, ABD­LDP­Ec demonstrated notable selective distribution in pancreatic carcinoma xenografts by passive targeting of albumin captured in the blood and was retained in the tumor for 48 h. ABD­LDP­Ec and ABD­LDP­Ec­AE exhibited inhibitory activity in cell proliferation and AsPC­1 xenograft growth, and ABD­LDP­Ec­AE increased the tumor growth inhibition rate by 20% compared with natural LDM. The results indicated that the introduction of ABD­based multi­functional drug delivery may be an effective approach to improve the efficacy of antitumor drugs, especially for K­ras mutant cancers.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Péptidos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Albúminas/química , Albúminas/genética , Aminoglicósidos/química , Aminoglicósidos/genética , Aminoglicósidos/farmacología , Animales , Antineoplásicos/química , Línea Celular Tumoral , Enediinos/química , Enediinos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Xenoinjertos , Humanos , Ratones , Mutación/genética , Oligopéptidos/química , Oligopéptidos/genética , Oligopéptidos/farmacología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Péptidos/genética , Unión Proteica/efectos de los fármacos , Dominios Proteicos/genética , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Phytomedicine ; 66: 153113, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31790901

RESUMEN

BACKGROUND: Natural killer (NK) cells play important roles in immune responses and have been wildly used in immunotherapy. Nevertheless, some limitations remain. It is urgent to explore novel and safe strategies to enhance NK cell activity. PURPOSE: The aim of this study was to investigate the immuno-stimulatory effects and to reveal the molecular mechanism of LJ101019C, a derivative of a natural small-molecule compounds cajanine, on NK cells. METHODS: Cell proliferation was examined by CCK8 assay, then we used the cytotoxicity detection kit to detect the cytotoxicity of NK cells. The change of cell cycle, intracellular reactive oxygen species (ROS) level and mitochondrial mass were evaluated by FACS and Operetta high-content image analysis, respectively. Furthermore, the IFN-γ secretion of NK cells were measured by ELISA. The Kv1.3 protein expression and function were detected by western blot and patch-clamp technique, respectively. The role of Kv1.3 in AKT/mTOR pathway activation was determined by western blot. RESULTS: The results showed that LJ101019C at relatively low concentrations (0.05-0.1 µM) significantly increased the proliferation of NK cells. And 1 µM LJ101019C could elevate the proportion of NK cells in the S-phase of the cell cycle (*p < 0.1). Furthermore, the cytotoxic effects of NK cells targeting MIA PaCa-2 cells were significantly enhanced by 0.1 and 1 µM LJ101019C, and were associated with the enhanced secretion of IFN-γ by NK cells (*p < 0.1; **p < 0.05). 0.1 and 1 µM LJ101019C increased intracellular levels of ROS (**p < 0.05), and 0.1 µM LJ101019C elevated mitochondrial mass (*p < 0.1). Electrophysiological recordings indicated that LJ101019C led to a remarkably increase the Kv1.3 current density. Moreover, western blot results indicated that LJ101019C elevated Kv1.3 protein expression and activated AKT/mTOR signaling via increasing the expression of Kv1.3 in NK cells. CONCLUSION: LJ101019C increases the proliferation and the cytotoxicity of NK cells at relatively low concentrations. The mechanism is the activation of AKT/mTOR signaling pathway driven by up-regulation of Kv1.3 in NK cells. These suggest LJ101019C is a promising candidate for improving the efficacy of NK cell-based immunotherapies.


Asunto(s)
Dietilestilbestrol/análogos & derivados , Canal de Potasio Kv1.3/efectos de los fármacos , Neoplasias/terapia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dietilestilbestrol/química , Dietilestilbestrol/farmacología , Femenino , Humanos , Inmunoterapia , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos BALB C , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
9.
Cancer Cell Int ; 19: 145, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31139022

RESUMEN

BACKGROUND: Pancreatic cancer stem cells (CSCs), a special population of cells, renew themselves infinitely and resist to various treatment. Gramicidin A (GrA), an ionophore antibiotic derived from microorganism, can form channels across the cell membrane and disrupt cellular ionic homeostasis, leading to cell dysfunction and death. As reported, the ionophore antibiotic salinomycin (Sal) has been proved to kill CSCs effectively. Whether GrA owns the potential as a therapeutic drug for CSCs still remains unknown. This study investigated the effect of GrA on pancreatic CSCs and the mechanism. METHODS: Tumorsphere formation assay was performed to assess pancreatic CSCs self-renewal potential. In vitro hemolysis assay was determined to test the borderline concentration of GrA. CCK-8 assay was used to detect pancreatic cancer cell proliferation capability. Flow cytometry was performed to detect cell apoptosis and mitochondrial membrane potential. Scanning and transmission electron microscopy was used to observe ultrastructural morphological changes on cell membrane surface and mitochondria, respectively. Western blot analysis was used to determine relative protein expression levels. Immunofluorescence staining was performed to observe CD47 re-distribution. RESULTS: GrA at 0.05 µM caused tumorspheres disintegration and decrease in number of pancreatic cancer BxPC-3 and MIA PaCa-2 cells. GrA and Sal both inhibited cancer cell proliferation. The IC50 values of GrA and Sal for BxPC-3 cells were 0.025 µM and 0.363 µM; while for MIA PaCa-2 cells were 0.032 µM and 0.163 µM, respectively. Compared on equal concentrations, the efficacy of GrA was stronger than that of Sal. GrA at 0.1 µM or lower did not cause hemolysis. GrA induced ultrastructural changes, such as the decrease of microvilli-like protrusions on cell surface membrane and the swelling of mitochondria. GrA down-regulated the expression levels of CD133, CD44, and CD47; in addition, CD47 re-distribution was observed on cell surface. Moreover, GrA showed synergism with gemcitabine in suppressing cancer cell proliferation. CONCLUSIONS: The study found that GrA was highly active against pancreatic CSCs. It indicates that GrA exerts inhibitory effects against pancreatic CSCs associated with CD47 down-regulation, implying that GrA might play a positive role in modulating the interaction between macrophages and tumor cells.

10.
Mol Pharm ; 15(6): 2403-2412, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29757658

RESUMEN

Enhanced macropinocytosis has been found in K-Ras mutant pancreatic cancer cells, through which albumin can massively enter into the K-Ras-driven cancer cells, suggesting its role in serving as a macropinocytosis-intensifying drug delivery carrier. In the present study, a novel recombinant protein Fv-LDP-D3 and its reconstituted analogue Fv-LDP-D3-AE were designed and prepared. Fv is the fragment of an anti-EGFR antibody, D3 is the domain III of human serum albumin (HSA), LDP is the apoprotein of the antitumor antibiotic lidamycin (LDM), and AE is an extremely cytotoxic enediyne chromophore derived from LDM. As shown, the recombinant protein Fv-LDP-D3 presented intensive and selective binding capacity to pancreatic cancer cells and inhibited cell proliferation by blocking EGFR signaling. Moreover, Fv-LDP-D3 showed prominent tumor imaging in pancreatic carcinoma xenograft. The reconstituted, enediyne-integrated analogue Fv-LDP-D3-AE displayed highly potent cytotoxicity to pancreatic cancer cells through apoptosis induction and G2/M arrest. Fv-LDP-D3 and Fv-LDP-D3-AE markedly inhibited the tumor growth of the pancreatic carcinoma AsPC-1 xenograft. Study results indicated that the novel recombinant protein displays both EGFR-targeting and macropinocytosis-intensifying attributes, presenting a new format of scFv antibody that integrates with albumin domain III. It might be a feasible strategy to develop targeted drugs for K-Ras mutant pancreatic cancer.


Asunto(s)
Antineoplásicos/farmacología , Inmunoconjugados/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Anticuerpos de Cadena Única/farmacología , Aminoglicósidos/química , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Enediinos/química , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Pinocitosis/efectos de los fármacos , Dominios Proteicos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Albúmina Sérica Humana/química , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
11.
PLoS One ; 13(2): e0191984, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29394294

RESUMEN

Through cell-based screening models, we have identified a new compound IMB5043, a thiophenylated pyridazinone, which exerted cytotoxicity against cancer cells. In the present study, we evaluated its antitumor efficacy and the possible mechanism. By MTT assay, IMB5043 inhibited the proliferation of various human cancer cells lines, especially hepatocarcinoma SMMC-7721 cells. IMB5043 blocked cell cycle with G2/M arrest, induced cell apoptosis, and inhibited the migration and invasion of SMMC-7721 cells. As verified by comet assay and γ-H2AX foci formation, IMB5043 caused DNA damage and activated ATM, Chk2 and p53 through phosphorylation. As shown by Gene microarray analysis, the differentially expressed genes in SMMC-7721 cells treated with IMB5043 were highly related to cell death and apoptosis. IMB5043 suppressed the growth of hepatocarcinoma SMMC-7721 xenograft in athymic mice. By histopathological examination, no lesions were found in bone marrow and various organs of the treated mice. Our findings reveal that IMB5043 as an active compound consisting of both pyridazinone and thiophene moieties exerts antitumor efficacy through activation of ATM-Chk2 pathway. IMB5043 may serve as a promising leading compound for the development of antitumor drugs.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Quinasa de Punto de Control 2/metabolismo , Piridazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Oncol Rep ; 38(6): 3507-3514, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29130101

RESUMEN

Previously it was shown that the recombinant EGFR/CD13 bi-targeted fusion protein ER(Fv)­LDP-NGR which consists of an anti­EGFR scFv antibody fragment, a tri­cyclic NGR peptide, and a lidamycin-derived apoprotein, inhibited the proliferation of cancer cells and markedly suppressed tumor growth of breast carcinoma MCF-7 xenografts in athymic mice. This study investigated the mechanism of action of the fusion protein. Human breast cancer MCF-7 cells, lung adenocarcinoma A549 cells, and microvascular endothelial HMEC­1 cells were used for a series of assays and determinations. ER(Fv)­LDP-NGR downregulated the transcription and expression of the target proteins EGFR and CD13, and interfered with the intracellular EGFR signaling pathway, cell cycle signaling pathway and apoptotic pathway. It induced apoptosis, inhibited proliferation, caused cell cycle G2/M phase arrest, and suppressed cell migration. Accompanied by weakening the capability to degrade extracellular matrix, ER(Fv)­LDP-NGR depressed the invasion capacity of cancer cells. In addition, ER(Fv)­LDP-NGR prevented microvascular endothelial cells from tube formation, which is closely related to angiogenesis. In conclusion, the EGFR/CD13 bi-targeted fusion protein ER(Fv)­LDP-NGR displays multi-functional characteristics, acting on both cancer cells and endothelial cells. It might be an effective agent for targeted cancer therapy.


Asunto(s)
Antígenos CD13/metabolismo , Regulación hacia Abajo , Receptores ErbB/metabolismo , Neoplasias/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Células A549 , Apoptosis , Antígenos CD13/genética , Ciclo Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptores ErbB/genética , Humanos , Células MCF-7 , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Oligopéptidos/genética , Oligopéptidos/metabolismo , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/efectos de los fármacos , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/metabolismo , Transcripción Genética/efectos de los fármacos
13.
Oncotarget ; 7(36): 58418-58434, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27517152

RESUMEN

K-Ras mutant pancreatic cancer cells display intensive macropinocytosis, indicating that this process may be exploited in the design of anticancer targeted therapies. In this study, we constructed a macropinocytosis-oriented recombinantly tailored defensin (DF-HSA) which consists of human ß-defensin-2 (DF) and human serum albumin (HSA). The macropinocytosis intensity and cytotoxicity of DF-HSA were investigated in K-Ras mutant MIA PaCa-2 cells and wild-type BxPC-3 cells. As found, the DF-HSA uptake in MIA PaCa-2 cells was much higher than that in wild-type BxPC-3 cells. Correspondingly, the cytotoxicity of DF-HSA to MIA PaCa-2 cells was more potent than that to BxPC-3 cells. In addition, the cytotoxicity of DF-HSA was much stronger than that of ß-defensin HBD2. DF-HSA suppressed cancer cell proliferation and induced mitochondrial pathway apoptosis. Notably, DF-HSA significantly inhibited the growth of human pancreatic carcinoma MIA PaCa-2 xenograft in athymic mice at well tolerated dose. By in vivo imaging, DF-HSA displayed a prominent accumulation in the tumor. The study indicates that the recombinantly tailored ß-defensin can intensively enter into the K-Ras mutant pancreatic cancer cells through macropinocytosis-mediated process and exert potent therapeutic efficacy against the pancreatic carcinoma xenograft. The novel format of ß-defensin may play an active role in macropinocytosis-mediated targeting therapy.


Asunto(s)
Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Pinocitosis , Proteínas Proto-Oncogénicas p21(ras)/genética , Albúmina Sérica Humana/metabolismo , beta-Defensinas/metabolismo , Células A549 , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Femenino , Genes ras , Vectores Genéticos , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo
14.
Basic Clin Pharmacol Toxicol ; 117(2): 105-16, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25615234

RESUMEN

Gelatinases play important roles in tumour invasion and metastasis and are thus considered promising targets for cancer therapy. In this study, a new single-chain variable fragment (scFv)-based fusion protein Fv-LDP, composed of the anti-gelatinases scFv and lidamycin apoprotein (LDP), was prepared, and its combination with angiogenesis inhibitor Endostar was then investigated. The fusion protein Fv-LDP specifically bound to various tumour cells, and its binding capability to human pulmonary giant cell carcinoma (PG) cells was higher than that of LDP. Fv-LDP inhibited the expression and secretion of gelatinases and could be internalized into tumour cells via endocytosis. Fv-LDP also suppressed the growth of human hepatoma cells and murine hepatoma 22 transplanted in Kunming mice in various degrees. In addition, Endostar could enhance the synergistic or additive inhibition of Fv-LDP on the growth, migration or invasion of human hepatoma cells shown by a colony formation assay and a transwell-based migration or invasion assay, respectively. In vivo, Fv-LDP/Endostar combination showed a significantly synergistic effect on the growth of a human hepatoma xenograft, with an inhibition rate of 80.8% compared with the Fv-LDP (44.1%) or Endostar (8.9%)-treated group. The above-mentioned results indicate that the fusion protein Fv-LDP is effective against transplantable hepatoma in mice and human hepatoma xenografts in athymic mice. Moreover, Endostar can potentiate the inhibition effect of Fv-LDP on the growth of human hepatoma cells and xenografts. These data will provide a new combined strategy for improving the therapeutic efficacy of treatments for hepatoma or other gelatinase-overexpressing tumours.


Asunto(s)
Aminoglicósidos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Endostatinas/farmacología , Enediinos/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Anticuerpos de Cadena Única/farmacología , Aminoglicósidos/administración & dosificación , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Apoproteínas/administración & dosificación , Apoproteínas/farmacología , Carcinoma de Células Gigantes/tratamiento farmacológico , Carcinoma de Células Gigantes/enzimología , Carcinoma de Células Gigantes/patología , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Endostatinas/administración & dosificación , Enediinos/administración & dosificación , Femenino , Gelatinasas/metabolismo , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Recombinantes , Anticuerpos de Cadena Única/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Anticancer Drugs ; 25(1): 82-91, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24100279

RESUMEN

Targeting to two or more objectives simultaneously has been pursued as a strategy to potentially increase the efficiency and selectivity of targeted drugs to certain cancers. In this study, an epidermal growth factor receptor (EGFR)/CD13-targeting, bispecific fusion protein ER(Fv)-LDP-NGR consisting of an anti-EGFR single-chain variable fragment (scFv), an apoprotein (LDP) of lidamycin (LDM), and a tri-CNGRC (Cys-Asn-Gly-Arg-Cys) peptide against CD13 was constructed, and then an enediyne-energized analog ER(Fv)-LDP-NGR-AE was generated by integration with an enediyne chomophore (AE) derived from LDM. The apoprotein LDP was used as a 'scaffold' to connect the scFv fragment and the tri-CNGRC peptide and also served as a specific 'carrier' for the extremely potent cytotoxic enediyne chromophore of LDM. Compared with its monospecific counterparts, ER(Fv)-LDP and LDP-NGR, the bispecific fusion protein ER(Fv)-LDP-NGR showed higher affinity to EGFR/CD13-overexpressed tumor cells. Determined by the MTT assay, the bispecific, enediyne-energized ER(Fv)-LDP-NGR-AE showed highly potent cytotoxicity to EGFR/CD13-overexpressed MCF-7 cells, with an IC50 value of 3.4×10 mol/l, whereas for the EGFR-overexpressed A431 cells, the IC50 value was 2.2×10 mol/l. For MCF-7 cells, the bispecific ER(Fv)-LDP-NGR-AE was more potent in cytotoxicity than the corresponding monospecific energized fusion proteins. In athymic mice models, the bispecific fusion protein ER(Fv)-LDP-NGR presented stronger inhibitory activity than the monospecific ER(Fv)-LDP and LDP-NGR. For the enediyne-energized fusion proteins, ER(Fv)-LDP-NGR-AE significantly inhibited the growth of EGFR/CD13-overexpressed MCF-7 xenograft and EGFR-overexpressed A431 xenograft by 86.3 and 81.4%, respectively. In addition, the bispecific ER(Fv)-LDP-NGR-AE showed much higher efficacy than its monospecific analogs ER(Fv)-LDP-AE and LDP-NGR-AE in both MCF-7 and A431 xenograft models. The results show that EGFR/CD13 bitargeting effectively improved the antitumor efficacy. Both the bispecific fusion protein and its enediyne-energized analog are highly effective in athymic mice bearing xenografts, and the latter exerts more marked efficacy. Generation of a pair of bispecific antibody-based therapeutics and its corresponding antibody-drug conjugate simultaneously may be a feasible strategy for the development of new targeted drugs for cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Antígenos CD13/metabolismo , Enediinos/uso terapéutico , Receptores ErbB/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Adenocarcinoma/tratamiento farmacológico , Animales , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Receptores ErbB/inmunología , Femenino , Humanos , Ratones , Ratones Desnudos , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Cadena Única/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Anticancer Drugs ; 23(4): 406-16, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22205154

RESUMEN

Epidermal growth factor receptor (EGFR), overexpressed in many epithelial tumors, plays important roles in the formation and the development of tumors, and thus it is regarded as a promising target for cancer therapy. Single-chain variable fragment (scFv), an engineered antibody fragment, is generally used for constructing antibody-targeted drugs, owing to its low immunogenicity and high penetration capability into solid tumors. A fusion protein ER(Fv-LDP), consisting of an anti-EGFR scFv and the apoprotein (LDP) of lidamycin (LDM), was prepared and then assembled with the active chomophore [active enediyne (AE)] of LDM to generate enediyne-energized analogue ER(Fv-LDP-AE). The fusion protein ER(Fv-LDP) bound specifically to EGFR-overexpressing cancer cells and internalized into the cytoplasm through receptor-mediated endocytosis. ER(Fv-LDP) possessed cytotoxicity against carcinoma cell lines, which was hundreds of times more potent than the separate moiety of ER(Fv) and LDP. The enediyne-energized fusion protein ER(Fv-LDP-AE) also showed stronger cytotoxicity to target-relevant cancer cells than LDM in vitro. In human epidermoid carcinoma A431 xenografts, ER(Fv-LDP) presented higher antitumor efficacy than that of ER(Fv), LDP, and their mixture, with tumor growth inhibition rates of 63.6, 46.7, 48.5, and 49.9%, respectively. The enediyne-energized fusion protein ER(Fv-LDP-AE) at a dose of 0.4 mg/kg inhibited tumor growth by 89.2%, while no significant body weight loss was seen in treated animals. The results show that an anti-EGFR scFv-based fusion protein and its enediyne-energized analogue can be prepared by DNA recombination and molecular reconstitution. Both ER(Fv-LDP) and ER(Fv-LDP-AE) are effective against EGFR-overexpressing cancer xenograft in athymic mice. An integrated technical platform for scFv-based enediyne-energized fusion proteins has been established.


Asunto(s)
Aminoglicósidos/farmacología , Apoproteínas/farmacología , Proliferación Celular/efectos de los fármacos , Enediinos/farmacología , Receptores ErbB/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Cadena Única/farmacología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Yao Xue Xue Bao ; 44(6): 597-602, 2009 Jun.
Artículo en Chino | MEDLINE | ID: mdl-19806889

RESUMEN

Recent studies have shown that epidermal growth factor receptor (EGFR) is an important target for cancer therapy. The present study prepared single chain Fv (scFv) directed against EGFR. Balb/c mice were immunized by human carcinoma A431 cells, and total RNA of the splenic cells was extracted. VH and VL gene fragments were amplified by RT-PCR and further joined into scFv gene with a linker, then scFv gene fragments were ligated into the phagemid vector pCANTAB 5E. The phagemid containing scFv were transformed into electro-competent E. coli TG1 cells. The recombinant phage antibody library was constructed through rescuing the transformed cells with help phage M13K07. The specified recombinant phages were enriched through 5 rounds of affinity panning and the anti-EGFR phage scFv clones were screened and identified with ELISA. A total of 48 clones from the library were selected randomly and 45 clones were identified positive. After infecting E. coli HB2151 cells with one positive clone, soluble recombinant antibodies about 27 kD were produced and located in the periplasm and the supernatant. The result of sequencing showed that the scFv gene was 768 bp, which encoded 256 amino acid residues. VH and VL including 3 CDRs and 4 FRs, respectively, were all homologous to mouse Ig. The soluble scFv showed the specific binding activity to purified EGFR and EGFR located in carcinoma cell membrane. The successful preparation of anti-EGFR scFv will provide an EGFR targeted molecule for the development of antibody-based drugs and biological therapy of cancer.


Asunto(s)
Receptores ErbB/genética , Cadenas Ligeras de Inmunoglobulina/genética , Anticuerpos de Cadena Única/genética , Animales , Anticuerpos Monoclonales , Línea Celular Tumoral , Receptores ErbB/inmunología , Humanos , Cadenas Ligeras de Inmunoglobulina/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Biblioteca de Péptidos , Anticuerpos de Cadena Única/inmunología
18.
Artículo en Chino | MEDLINE | ID: mdl-16222100

RESUMEN

The recombinant plasmid pBIBSa containing the HBsAg DNA fragment was transferred into Agrobacterium tumefaciens strain LBA4404 directly. Ginseng cells were co-cultivated with Agrobacterium tumefaciens carrying pBIBSa. The ginseng cell lines carrying HBsAg-S gene were obtained. Transgenic cells were checked for the presence of target gene using PCR and RT-PCR. Samples containing the target gene showed a clear band at the site of 700 bp by agarose electrophoresis analysis (Figs. 2, 3). Expression levels determined by ELISA showed maximum expression levels of 184 ng HBsAg/g FW and 0.009% of the total soluble protein. HBsAg in ginseng cells were located both on the membrane and in the nuclei (Fig. 4).


Asunto(s)
Antígenos de Superficie de la Hepatitis B/genética , Panax/genética , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Antígenos de Superficie de la Hepatitis B/metabolismo , Panax/citología , Panax/metabolismo , Plantas Modificadas Genéticamente/citología , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/metabolismo , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA