Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Int J Hyperthermia ; 39(1): 405-413, 2022.
Article in English | MEDLINE | ID: mdl-35236209

ABSTRACT

BACKGROUND: Enediynes are anti-cancer agents that are highly cytotoxic due to their propensity for low thermal activation of radical generation. The diradical intermediate produced from Bergman cyclization of the enediyne moiety may induce DNA damage and cell lethality. The cytotoxicity of enediynes and difficulties in controlling their thermal cyclization has limited their clinical use. We recently showed that enediyne toxicity at 37 °C can be mitigated by metallation, but cytotoxic effects of 'metalloenediynes' on cultured tumor cells are potentiated by hyperthermia. Reduction of cytotoxicity at normothermia suggests metalloenediynes will have a large therapeutic margin, with cell death occurring primarily in the heated tumor. Based on our previous in vitro findings, FeSO4-PyED, an Fe co-factor complex of (Z)-N,N'-bis[1-pyridin-2-yl-meth-(E)-ylidene]oct-4-ene-2,6-diyne-1,8-diamine, was prioritized for further in vitro and in vivo testing in normal human melanocytes and melanoma cells. METHODS: Clonogenic survival, apopotosis and DNA binding assays were used to determine mechanisms of enhancement of FeSO4-PyED cytotoxicity by hyperthermia. A murine human melanoma xenograft model was used to assess in vivo efficacy of FeSO4-PyED at 37 or 42.5 °C. RESULTS: FeSO4-PyED is a DNA-binding compound. Enhancement of FeSO4-PyED cytotoxicity by hyperthermia in melanoma cells was due to Bergman cyclization, diradical formation, and increased apoptosis. Thermal enhancement, however, was not observed in melanocytes. FeSO4-PyED inhibited tumor growth when melanomas were heated during drug treatment, without inducing normal tissue damage. CONCLUSION: By leveraging the unique thermal activation properties of metalloenediynes, we propose that localized moderate hyperthermia can be used to confine the cytotoxicity of these compounds to tumors, while sparing normal tissue.


Subject(s)
Antineoplastic Agents , Hyperthermia, Induced , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cyclization , Enediynes/chemistry , Enediynes/pharmacology , Enediynes/therapeutic use , Hot Temperature , Humans , Mice
2.
Biol Pharm Bull ; 44(7): 1024-1028, 2021.
Article in English | MEDLINE | ID: mdl-34193685

ABSTRACT

Brain inflammation is a pathological characteristic of neurodegenerative diseases. In this condition, excessively activated microglia elevate proinflammatory mediator levels. We previously reported that panaxytriol inhibited lipopolysaccharide (LPS)-induced microglia activation in vitro. However, the effects of panaxytriol on microglia activation in vivo require confirmation. In the present study, we found that panaxytriol suppressed both microglia and astrocyte activation by injected LPS intracerebrally to mice with LPS-induced brain inflammation. Panaxytriol was more effective on microglia than astrocytes. Moreover, panaxytriol tended to reduce LPS-induced spontaneous motor activity dysfunction. These results suggested that panaxytriol could improve brain health by suppressing microglia activation in neurodegenerative diseases.


Subject(s)
Encephalitis/drug therapy , Enediynes/therapeutic use , Fatty Alcohols/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Astrocytes/drug effects , Enediynes/pharmacology , Fatty Alcohols/pharmacology , Hippocampus/drug effects , Lipopolysaccharides/pharmacology , Locomotion/drug effects , Mice, Inbred C57BL , Microglia/drug effects , Neuroprotective Agents/pharmacology
3.
Oncol Rep ; 41(1): 475-482, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30542729

ABSTRACT

Apoptosis induction and differentiation of promyelocytic leukemic cells into mature cells are major strategies for the drug-based treatment of leukemia. Lidamycin (LDM) which is a member of the enediyne antibiotic family exhibits extreme cytotoxicity. In the present study, the induction of apoptosis and differentiation in human chronic myeloid leukemia K562 cells by low concentrations of lidamycin were investigated. K562 cells were treated with lidamycin at various concentrations for 48 h, and accumulated in the metaphase as determined in previous experiments. Cell viability was determined using a Cell Counting Kit-8 (CCK-8) assay and the IC50 value of lidamycin was 0.1±3.2 nM. Induction of apoptosis was investigated morphologically by acridine orange/ethidium bromide (AO/EtBr) staining. Growth inhibition and apoptosis induction were observed in cells treated with low concentrations of lidamycin. In addition, western blot analysis revealed that treatment of the K562 cells with lidamycin at low concentrations upregulated the expression of caspase-8 and caspase-3. The induction of differentiation in human chronic myeloid leukemia K562 cells by lidamycin at low concentrations was also investigated. The nitroblue tetrazolium reduction ability of K562 cells was increased following treatment with lidamycin. Low concentrations of lidamycin triggered erythroid differentiation among K562 cells, indicated by morphological changes, increased hemoglobin content, and the expression of cell surface antigens such as CD71. Additionally the expression of GATA-binding factor 1 (GATA-1) protein in low concentration lidamycin-treated K562 cells was increased. The results of the present study suggest that a low-concentration lidamycin exerts effects on apoptosis and erythroid differentiation induction by increasing the expression of caspases and GATA-1 protein. Lidamycin may serve a positive role in relevant targeted chemotherapy and may represent a potential candidate for chronic myelogenous leukemia differentiation-inducing treatment.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Enediynes/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Aminoglycosides/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antigens, CD/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enediynes/therapeutic use , GATA1 Transcription Factor/metabolism , Hemoglobins/metabolism , Humans , K562 Cells , Nitroblue Tetrazolium/metabolism , Receptors, Transferrin/metabolism
4.
Bioconjug Chem ; 29(9): 3104-3112, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30105903

ABSTRACT

Depending on increasing extracellular protein utilization and altering metabolic programs, cancer cells could proliferate and survive without restricion by ingesting human serum albumin (HSA) to serve as nutritional amino acids. Here, we hypothesize that the consumption of albumin by cancer cells could be utilized as an efficient approach to targeted drug delivery. Lidamycin (LDM), an antitumor antibiotic with extremely potent cytotoxicity to cultured cancer cells, consists of an apoprotein (LDP) and an active enediyne chromophore (AE). In the present study, a novel albumin-lidamycin conjugate was prepared by DNA recombination and molecular reconstitution. Results show that the IC50 values of albumin-lidamycin conjugate (HSA-LDP-AE) for a variety of tested cancer cells were at subnanomolar levels. At tolerated doses, the albumin-lidamycin conjugate significantly inhibited the growth of lung carcinoma PG-BE1 xenografts by 97.8%. The therapeutic efficacy of the albumin-lidamycin conjugate was much stronger than that of free lidamycin. Meanwhile, the images of albumin-lidamycin conjugate showed obvious and lasting tumor localization and fluorescence enrichment and there was no detectable signal in nontumor locations. Taken together, albumin-lidamycin conjugate, a new format of lidamycin, could be a promising antitumor therapeutic agent and albumin-integration might be a feasible approach to targeted antitumor drug delivery.


Subject(s)
Albumins/chemistry , Aminoglycosides/chemistry , Aminoglycosides/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Enediynes/chemistry , Enediynes/therapeutic use , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Female , Humans , Mice , Optical Imaging , Xenograft Model Antitumor Assays
5.
Acta Pharmacol Sin ; 39(11): 1777-1786, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30013033

ABSTRACT

Defensins play an essential role in innate immunity. In this study, a novel recombinant ß-defensin that targets the epidermal growth factor receptor (EGFR) was designed and prepared. The EGFR-targeting ß-defensin consists of an EGF-derived oligopeptide (Ec), a ß-defensin-1 peptide (hBD1) and a lidamycin-derived apoprotein (LDP), which serves as the "scaffold" for the fusion protein (Ec-LDP-hBD1). Ec-LDP-hBD1 effectively bound to EGFR highly expressed human epidermoid carcinoma A431 cells. The cytotoxicity of Ec-LDP-hBD1 to EGFR highly expressed A431 cells was more potent than that to EGFR low-expressed human lung carcinoma A549 and H460 cells (the IC50 values in A431, A549, and H460 cells were 1.8 ± 0.55, 11.9 ± 0.51, and 5.19 ± 1.21 µmol/L, respectively); in addition, the cytotoxicity of Ec-LDP-hBD1 was much stronger than that of Ec-LDP and hBD1. Moreover, Ec-LDP-hBD1 suppressed cancer cell proliferation and induced mitochondria-mediated apoptosis. Its in vivo anticancer action was evaluated in athymic mice with A431 and H460 xenografts. The mice were administered Ec-LDP-hBD1 (5, 10 mg/kg, i.v.) two times with a weekly interval. Administration of Ec-LDP-hBD1 markedly inhibited the tumor growth without significant body weight changes. The in vivo imaging further revealed that Ec-LDP-hBD1 had a tumor-specific distribution with a clear image of localization. The results demonstrate that the novel recombinant EGFR-targeting ß-defensin Ec-LDP-hBD1 displays both selectivity and enhanced cytotoxicity against relevant cancer cells by inducing mitochondria-mediated apoptosis and exhibits high therapeutic efficacy against the EGFR-expressed carcinoma xenograft. This novel format of ß-defensin, which induces mitochondrial-mediated apoptosis, may play an active role in EGFR-targeting cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Mitochondria/metabolism , Recombinant Fusion Proteins/therapeutic use , beta-Defensins/therapeutic use , Aminoglycosides/metabolism , Aminoglycosides/therapeutic use , Animals , Antineoplastic Agents/metabolism , Apoproteins/metabolism , Apoproteins/therapeutic use , Cell Line, Tumor , Enediynes/metabolism , Enediynes/therapeutic use , ErbB Receptors/metabolism , Female , Humans , Mice, Nude , Mitochondria/pathology , Protein Binding , Recombinant Fusion Proteins/metabolism , Xenograft Model Antitumor Assays , beta-Defensins/metabolism
6.
Mol Pharm ; 15(3): 768-797, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29303588

ABSTRACT

Spatial and temporal control over DNA cleavage by photoactivated enediynes can be complemented by additional factors such as the release of internal strain, chelation, pH changes, intramolecular H-bonds, and substituent effects. This review presents design and reactivity of photoactivated enediynes/enynes and analyses the chemical, biological, and photophysical challenges in their applications.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA, Neoplasm/drug effects , Enediynes/pharmacology , Neoplasms/drug therapy , Photochemotherapy/methods , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/radiation effects , Antibiotics, Antineoplastic/therapeutic use , Cyclization/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA, Neoplasm/chemistry , DNA, Neoplasm/radiation effects , Enediynes/chemistry , Enediynes/radiation effects , Enediynes/therapeutic use , Humans , Light , Lysine/chemistry , Molecular Structure , Molecular Targeted Therapy/methods , Neoplasms/genetics
7.
Curr Med Chem ; 24(32): 3433-3484, 2017.
Article in English | MEDLINE | ID: mdl-28443507

ABSTRACT

BACKGROUND: The first reports of the natural enediyne anticancer antibiotics date back to the late 1980s; since then, a great deal of interest has been devoted to the chemistry, biology and potential medical applications of this family of compounds. The biological activity of enediynes is linked to the presence of a highly unsaturated hex-1-ene-1,5-diyne system. The thermally induced transformation of this unit into a benzene σ-σ diradical (the Bergman cycloaromatization) is the key step of the antitumor properties of such compounds: 1,4-diaryl radicals are able to abstract H-atoms from the deoxyribose backbone of DNA, thus leading to DNA strand cleavage and ultimately cell death. METHODS: We undertook a structured search of bibliographic databases for peer-reviewed research literature using focused and high quality papers. Research efforts addressed at understanding and mimicking the various processes involved in the targeting, activation and DNA cleavage associated with these products are described. The potential of a great number of non natural enediynes in the treatment of many infectious diseases, apart their role in anticancer drugs, such as antibacterial activity, protein degradation activity, has been reported Results: Due to the interesting mode of action of this class of compounds, the unique molecular architecture of enediynes has been exploited towards the synthesis of many non natural compounds in order to study and enhance their biological properties. Seventy-six papers were included in this review. It is divided in paragraphs that include: Carbo- oxygen-nitrogen- and sulfur- enediynes, polymers and macrocycles. The synthetic approaches to the different classes of compounds are discussed in detail together with the biological implications of the synthesized compounds Conclusion: The review summarizes the most recent advances in the synthesis and reactivity of non natural enediynes by focusing the attention particularly to the biological properties of the most interesting members of the family of carbo- and hetero- enediynes. The findings of this review confirm the importance of non natural enediynes as potential drugs in the treatment of cancer and many infectious diseases.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Chemistry Techniques, Synthetic/methods , Enediynes/chemical synthesis , Enediynes/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA Cleavage/drug effects , Enediynes/chemistry , Enediynes/pharmacology , Humans , Neoplasms/genetics
8.
PLoS One ; 10(12): e0143206, 2015.
Article in English | MEDLINE | ID: mdl-26659358

ABSTRACT

RESEARCH BACKGROUND: Currently, multiple myeloma is the second most common hematological malignancy in the U.S., constituting 1% of all cancers. With conventional treatment, the median survival time is typically 3-4 years, although it can be extended to 5-7 years or longer with advanced treatments. Recent research indicated that an increase in osteoclast (OC) activity is often associated withmultiple myeloma (MM) and that a decrease inosteoblast (OB) activity contributesto the osteolytic lesions in MM. Normally, the populations of OCs and OBs are inequilibrium, and an imbalance in this statecontributes to the development of lesions. RESEARCH PROCEDURES: A multi-scale agent-based multiple myeloma model was developed to simulate the proliferation, migration and death of OBs and OCs. Subsequently, this model was employed to investigate the efficacy of thethree most commonly used drugs for MM treatment under the following two premises: the reduction in the progression of MM and the re-establishment of the equilibrium between OCs and OBs. RESEARCH PURPOSES: The simulated results not only demonstrated the capacity of the model to choose optimal combinations of the drugs but also showed that the optimal use of the three drugs can restore the balance between OCs and OBs as well as kill MMs. Furthermore, the drug synergism analysis function of the model revealed that restoring the balance between OBs and OCs can significantly increase the efficacy of drugs against tumor cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Models, Biological , Models, Statistical , Multiple Myeloma/drug therapy , Osteoblasts/drug effects , Osteoclasts/drug effects , Aminoglycosides/pharmacokinetics , Aminoglycosides/therapeutic use , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Enediynes/pharmacokinetics , Enediynes/therapeutic use , Gene Expression Regulation, Neoplastic , Glucocorticoids/pharmacokinetics , Glucocorticoids/therapeutic use , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteoblasts/pathology , Osteoclasts/pathology , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Wnt Signaling Pathway
9.
DNA Repair (Amst) ; 21: 165-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24986640

ABSTRACT

The ability of the radiomimetic anti-tumor enediyne C-1027 to induce DNA inter-strand crosslinks (ICLs), in addition to the expected DNA strand breaks, is unique among traditional DNA targeted cancer therapies. Importantly, radiation therapy and most radiomimetic drugs have diminished effect in hypoxic environments due to decreased induction of DNA strand breaks, which is an oxygen requiring process. However, C-1027's induction of ICLs is enhanced under hypoxia and it is actually more potent against hypoxic cells, overcoming this common tumor resistance mechanism. In this study, an analog of C-1027, 20'-deschloro-C-1027 was examined for its ability to induce DNA ICLs under hypoxic conditions. Deschloro-induced ICLs were detected under hypoxic cell-free conditions, with a concomitant reduction in the induction of DNA strand breaks. In cells deschloro behaved similarly, inducing cellular ICLs under hypoxic conditions with a reduction in DNA breaks. The cytotoxicity of deschloro treatment was similar in normoxic and hypoxic cells, suggesting that the ICL induction allows deschloro to retain its cytotoxic activity under hypoxia. It appears that rational engineering of the C-1027 family of radiomimetics holds promise toward overcoming the radioresistance associated with the hypoxic environment associated with solid tumors.


Subject(s)
Aminoglycosides/toxicity , Antibiotics, Antineoplastic/toxicity , Cross-Linking Reagents/toxicity , DNA Breaks , Enediynes/toxicity , Radiation Tolerance , Aminoglycosides/therapeutic use , Cell Hypoxia , Enediynes/therapeutic use , HCT116 Cells , Humans
10.
J Neurooncol ; 119(1): 91-100, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24842385

ABSTRACT

The present work evaluated the synergistic efficacy of an enediyne antibiotic lidamycin (LDM) plus temozolomide (TMZ) against glioma in vitro and in vivo. LDM plus TMZ inhibited the proliferations of rat glioma C6 cells and human glioma U87 cells more efficiently than the single usage of LDM or TMZ. In addition, LDM also potentiated the apoptosis inductions by TMZ in rat C6 cells and human U87 cells. Meanwhile, the results of TdT-mediated dUTP Nick End Labeling assay for subcutaneous U87 tumor sections indicated an enhanced apoptosis induction in vivo by LDM plus TMZ, which confirmed the high potency of the combination for glioma therapy. As determined by Western blot, apoptosis signal pathways in C6 cells and U87 cells were markedly affected by the synergistic alteration of P53, bax, procaspase 3, and bcd-2 expression. In both subcutaneous U87 xenograft and C6 intracerebral orthotopic implant model, TMZ-induced glioma growth suppression was dramatically potentiated by LDM. As shown, the combination therapy efficiently reduced the tumor volumes and tumor weights of the human glioma U87 xenograft. Kaplan-Meier assay revealed that LDM plus TMZ dramatically prolonged the life span of C6 intracerebral tumor-bearing rats with decreased tumor size. This study indicates that the combination of LDM with TMZ might be a promising strategy for glioma therapy.


Subject(s)
Aminoglycosides/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Enediynes/therapeutic use , Glioma/drug therapy , Aminoglycosides/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Synergism , Enediynes/pharmacology , Glioma/pathology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Rats , Signal Transduction/drug effects , Temozolomide
11.
Cancer Biol Ther ; 15(4): 398-408, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24424202

ABSTRACT

Present work mainly evaluated the inhibitory effects of lidamycin (LDM), an enediyne antibiotic, on angiogenesis or glioma-induced angiogenesis in vitro and in vivo, especially its synergistic anti-angiogenesis with temozolomide (TMZ). LDM alone efficiently inhibited proliferations and induced apoptosis of rat brain microvessel endothelial cells (rBMEC). LDM also interrupted the tube formation of rat brain microvessel endothelial cells (rBMEC) and rat aortic ring spreading. The blockade of rBMEC invasion and C6 cell-induced rBMEC migration by LDM was associated with decrease of VEGF secretion in a co-culture system. TMZ dramatically potentiated the effects of LDM on anti-proliferation, apoptosis induction, and synergistically inhibited angiogenesis events. As determined by western blot and ELISA, the interaction of tumor cells and the rBMEC was markedly interrupted by LDM plus TMZ with synergistic regulations of VEGF induced angiogenesis signal pathway, tumor cell invasion/migration, and apoptosis signal pathway. Immunofluorohistochemistry of CD31 and VEGF showed that LDM plus TMZ resulted in synergistic decrease of microvessel density (MVD) and VEGF expression in human glioma U87 cell subcutaneous xenograft. This study indicates that the high efficacy of LDM and the synergistic effects of LDM plus TMZ against glioma are mediated, at least in part, by the potentiated anti-angiogenesis.


Subject(s)
Aminoglycosides/pharmacology , Angiogenesis Inhibitors/pharmacology , Anti-Bacterial Agents/pharmacology , Brain Neoplasms/blood supply , Dacarbazine/analogs & derivatives , Enediynes/pharmacology , Glioma/blood supply , Neovascularization, Pathologic/metabolism , Aminoglycosides/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Apoptosis/drug effects , Brain/blood supply , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Synergism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enediynes/therapeutic use , Glioma/drug therapy , Glioma/pathology , Heterografts , Humans , Male , Mice, Nude , Rats , Temozolomide , Vascular Endothelial Growth Factor A/metabolism
12.
Anticancer Drugs ; 25(1): 82-91, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24100279

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , CD13 Antigens/metabolism , Enediynes/therapeutic use , ErbB Receptors/genetics , Recombinant Fusion Proteins/therapeutic use , Adenocarcinoma/drug therapy , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , ErbB Receptors/immunology , Female , Humans , Mice , Mice, Nude , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/genetics , Xenograft Model Antitumor Assays
13.
Int J Mol Med ; 29(3): 409-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22108624

ABSTRACT

The overexpression of tissue factor (TF) observed in numerous cancer cells and clinical samples of human cancers make TF an ideal target for cancer therapy. Here, we report an energized fusion protein, hlFVII-LDP-AE, which can be used for cancer therapy and is composed of a human Factor VII light chain (hlFVII) conjugated to the cytotoxic antibiotic lidamycin (LDM, LDP-AE). hlFVII-LDP-AE binds with specificity to TF expressed on tumor cells, resulting in internalization of the fusion protein and cytotoxicity induced by the LDM domain. The potential efficacy of hlFVII-LDP-AE for cancer therapy was examined in vitro by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and in vivo with a BALB/c nude mouse xenograft model of the human lung cancer line NCI-H292. hlFVII-LDP-AE caused chromatin condensation and cleavage of genomic DNA in NCI-H292 cells. In the MTT assays, the IC50 value of hlFVII- LDP-AE was 0.19 nM. In the in vivo tests, after two intravenous injections of hlFVII-LDP-AE at a dose of 0.6 mg/kg, the growth rate of the lung tumor xenograft was reduced to 15% of the control rate, and there was no excessive loss of body weight and inflammatory response in the mice. These findings suggest that hlFVII-LDP-AE is efficacious and tolerated in the mouse model of NCI-H292 human lung cancer examined and could have broad clinical applicability for treating cancer patients.


Subject(s)
Adenocarcinoma/drug therapy , Aminoglycosides/genetics , Aminoglycosides/therapeutic use , Antineoplastic Agents/therapeutic use , Enediynes/therapeutic use , Factor VII/genetics , Lung Neoplasms/drug therapy , Recombinant Fusion Proteins/therapeutic use , Thromboplastin/metabolism , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Aminoglycosides/metabolism , Aminoglycosides/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly/drug effects , DNA Damage/drug effects , Enediynes/metabolism , Enediynes/pharmacology , Factor VII/metabolism , Female , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Binding , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Thromboplastin/genetics , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem Lett ; 20(24): 7549-52, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21050753

ABSTRACT

We synthesized diyne triols based on gymnasterkoreayne and evaluated their cancer chemopreventive activities in terms of the chemopreventive index (CI) to reveal the structure-activity relationship, and discovered more active compounds than natural diynes.


Subject(s)
Acetylene/analogs & derivatives , Anticarcinogenic Agents/chemistry , Enediynes/chemistry , Acetylene/chemical synthesis , Acetylene/chemistry , Acetylene/therapeutic use , Anticarcinogenic Agents/chemical synthesis , Anticarcinogenic Agents/therapeutic use , Enediynes/chemical synthesis , Enediynes/therapeutic use , Humans , Structure-Activity Relationship
15.
Gene Ther ; 17(10): 1234-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20463754

ABSTRACT

Lidamycin (LDM) is a new member of enediyne antitumor antibiotics family that can be separated and reconstituted. It consists of a labile active enediyne chromophore (AE) and a noncovalently bound apoprotein (LDP). LDM is now in phase II clinical trials. In this study, we described the antitumor features of a fusion protein of LDM, anti-CD20Fab-LDM, targeted to CD20 expressed by B-lymphoid malignancies. Especially, LDM was prepared by a novel two-step method including DNA recombination and molecular reconstitution. Anti-CD20Fab-LDM exerted potent cytotoxicity against CD20+ B-cell lymphoma cell lines in vitro (IC50: 10-30 pM) and in the Raji xenograft model. Two Raji xenografts were allowed to grow to an initial mass of 80 and 500 mm³, respectively, and then anti-CD20Fab-LDM was administered intravenously with the highest dose of 4 nmol kg⁻¹ . The inhibition rates of tumor growth were 90.1 and 85%, which were saliently superior to those of nontargeted LDM. It is noteworthy that anti-CD20Fab-LDM can inhibit the growth of patient-derived cells, including rituximab-resistant patient-derived cells. Thus, CD20-targeted delivery of LDM is a specific and potent therapeutic strategy for B-lymphoid malignancies. In addition, the two-step approach could serve as a new technology platform for making a series of highly potent engineered antibody-based drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, B-Cell/drug therapy , Recombinant Fusion Proteins/therapeutic use , Aminoglycosides/chemistry , Aminoglycosides/therapeutic use , Animals , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Monoclonal, Murine-Derived/metabolism , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Line, Tumor , Enediynes/chemistry , Enediynes/therapeutic use , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transplantation, Heterologous
16.
Cancer Lett ; 295(1): 124-33, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20303650

ABSTRACT

Gelatinases play important roles in tumor progression and are abundantly expressed in a variety of malignant tumors. Antibody targeting gelatinases is a possible avenue to fight against cancer. However, antibody alone can not achieve curative efficacy. Herein, we demonstrated the intensified targeting therapy of a tandem scFv-based fusion protein and its enediyne-energized analogue against gelatinases-overexpressed tumor. A fusion protein dFv-LDP, comprising a tandem scFv of anti-gelatinases linked to the apoprotein (LDP) of lidamycin, was generated and showed strong tumor targeting capability in three different tumor xenografts. In PG-BE1 lung carcinoma xenograft, the tumor inhibition rate was 77.5% by dFv-LDP versus 94.2% by dFv-LDP-AE, the product of dFv-LDP assembled with the active enediyne chromophore (AE) of lidamycin. Moreover, the combination of dFv-LDP with dFv-LDP-AE further augmented the therapeutic efficacy, producing initial tumor shrinkage in five of six mice. The microvessel density (P<0.05) and proliferation index (P<0.05) were also stepwise decreased in groups of dFv-LDP, dFv-LDP-AE and the combination. In conclusion, our results demonstrated that the antibody-based therapy against gelatinases was stepwise intensified in use of dFv-LDP, dFv-LDP-AE and dFv-LDP plus dFv-LDP-AE, and indicated that the combination of an antibody with its drug-armed analogue might be of interest as a new approach to augment antitumor efficacy.


Subject(s)
Aminoglycosides/therapeutic use , Antineoplastic Agents/therapeutic use , Apoproteins/therapeutic use , Enediynes/therapeutic use , Gelatinases/immunology , Lung Neoplasms/drug therapy , Single-Chain Antibodies/therapeutic use , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Cell Proliferation/drug effects , Enediynes/chemistry , Enediynes/metabolism , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Mice , Mice, Nude , Microvessels/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , Xenograft Model Antitumor Assays
17.
Clin Cancer Res ; 16(7): 2085-94, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20332319

ABSTRACT

PURPOSE: The cooverexpression of epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) observed in many human tumors and their synergistic interaction in the transformation of cells make these receptors important targets for the development of new targeted therapeutics. Targeting of EGFR and HER2 simultaneously has been pursued as a strategy with which to potentially increase efficiency and selectivity in therapy of certain cancers. This study was set to construct a bispecific energized fusion protein (Ec-LDP-Hr-AE) consisting of two oligopeptides against EGFR and HER2, and lidamycin, and investigate its antitumor efficacy. EXPERIMENTAL DESIGN: In vitro experiments measured the binding and internalization of bispecific Ec-LDP-Hr fusion protein. The potency of energized fusion proteins was also done in which the bispecific Ec-LDP-Hr-AE was compared with lidamycin (LDM) and its monospecific counterparts, Ec-LDP-AE and LDP-Hr-AE. In vivo, Ec-LDP-Hr-AE was given i.v. to nude mice bearing human ovarian carcinoma SK-OV-3 xenografts. RESULTS: Binding and internalization studies showed that bispecific fusion protein Ec-LDP-Hr bound to carcinoma cells specifically and then were internalized into the cytoplasm. Bispecific Ec-LDP-Hr-AE was more potent and selective in its cytotoxicity against different carcinoma cell lines than corresponding momospecific agents and LDM in vitro. In addition, Ec-LDP-Hr-AE significantly inhibited the growth of SK-OV-3 xenografts in nude mouse model. In vivo imaging study showed that FITC-labeled Ec-LDP-Hr was targeted and accumulated in the tumors. CONCLUSION: A ligand-based and an antibody-based oligopeptide fused to the enediyne antibiotic LDM created a new bispecific fusion protein with low molecular weight and more potent in vitro and in vivo antitumor activity (than momospecific fusion proteins).


Subject(s)
Antineoplastic Agents/therapeutic use , Enediynes/chemistry , ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/pharmacology , Antibodies/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enediynes/immunology , Enediynes/pharmacology , Enediynes/therapeutic use , ErbB Receptors/immunology , Female , Humans , Ligands , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms/pathology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Substrate Specificity , Treatment Outcome , Xenograft Model Antitumor Assays
18.
Bioconjug Chem ; 20(8): 1587-94, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19572629

ABSTRACT

Children with relapsed and refractory acute lymphoblastic leukemia (ALL) still face a critical prognosis. We tested the hypothesis that targeted calicheamicin theta (θ) using an anti-CD19-immunoconjugate may provide an effective treatment strategy for CD19(+) ALL. Calicheamicin θ is a rationally designed prodrug of the natural enediyene calicheamicin γ, obtained by total synthesis. It offers the advantage of increased in vivo stability and 1000-fold higher antitumor potency over calicheamicin γ. First, we demonstrate efficacy of calicheamicin θ against primary pre-B leukemic cells and multidrug-resistant leukemia cell lines (IC(50) = 10(-9) to 10(-12) M). Second, conjugation of calicheamicin θ to an internalizing murine anti-CD19 monoclonal antibody was demonstrated to affect neither calicheamicin θ mediated cytotoxicity nor binding of the antibody to the target molecule. Third, anti-CD19-calicheamicin θ immunoconjugate revealed a maximum tolerated dose of 10 µg/kg and CD19-specific and long-lasting eradication of established leukemia was demonstrated in a xenograft model. Finally, we show that the antileukemic effect of anti-CD19-calicheamicin θ is mediated by induction of apoptosis proceeding through the caspase-mediated mitochondrial pathway. On the basis of these results, we conclude that anti-CD19-calicheamicin θ immunoconjugates may offer a novel and effective approach for the treatment of relapsed CD19(+) ALL.


Subject(s)
Aminoglycosides/therapeutic use , Antigens, CD19/metabolism , Antineoplastic Agents/therapeutic use , Enediynes/therapeutic use , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Aminoglycosides/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antigens, CD19/immunology , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enediynes/chemistry , Female , Humans , Immunoconjugates/immunology , Mice , Mice, SCID , Molecular Structure , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
19.
Acta Pharmacol Sin ; 30(7): 1025-32, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19575006

ABSTRACT

AIM: To investigate the effects of lidamycin (LDM) on a mouse myeloma cell line (SP2/0) and human multiple myeloma cell lines (U266 and SKO-007), and provide the basis for the use of LDM in cancer therapy. METHODS: A 3-[4,5-dimethylthiazol-2-yl]5-[3-carboxymethoxyphenyl]-2-[4-sulfophenyl]2H-tetrazolium inner salt (MTS) assay was used to determine the degree of growth inhibition by the drugs analyzed in this study. Cell cycle distribution and analysis were measured by flow cytometry combined with propidium iodide (PI) staining. The effects on apoptosis were measured by Hoechst 33342 staining and by flow cytometry combined with fluorescein-isothiocyanate-Annexin V/propidium iodide (FITC-Annexin V/PI) staining. Protein expression was determined by Western blot analysis. In vivo antitumor activity was measured using a murine myeloma model in BALB/c mice. RESULTS: There was a significant reduction in cell proliferation after treatment with LDM. The overall growth inhibition correlated with increased apoptotic cell death. LDM-induced cell apoptosis was associated with the activation of c-Jun-N-terminal kinase (JNK), and cleavage of caspase-3/7 and poly (ADP-ribose) polymerase (PARP). LDM markedly suppressed tumor growth in a murine myeloma model. CONCLUSION: LDM induces apoptosis in murine myeloma SP2/0 cells as well as in human myeloma U266 and SKO-007 cell lines. The sustained activation of JNK might play a critical role in LDM-induced apoptosis in the SP2/0 cell line. LDM demonstrates significant antitumor efficacy against myeloma SP2/0 cells in mice. Taken together, our data provide some clues for further research of the effects of LDM on human multiple myeloma.Acta Pharmacologica Sinica (2009) 30: 1025-1032; doi: 10.1038/aps.2009.75.


Subject(s)
Aminoglycosides , Antibiotics, Antineoplastic , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Enediynes , Multiple Myeloma , Aminoglycosides/pharmacology , Aminoglycosides/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/physiology , Enediynes/pharmacology , Enediynes/therapeutic use , Enzyme Activation , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred BALB C , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neoplasm Transplantation , Random Allocation
20.
Int J Hematol ; 90(1): 44-51, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19468799

ABSTRACT

In the present study, the effects of lidamycin (LDM), a member of the enediyne antibiotic family, on two human multiple myeloma (MM) cell lines, U266 and SKO-007, were evaluated. In MTS assay, LDM showed much more potent cytotoxicity than conventional anti-MM agents to both cell lines. The IC(50) values of LDM for the U266 and SKO-007 cells were 0.0575 +/- 0.0015 and 0.1585 +/- 0.0166 nM, respectively, much lower than those of adriamycin, dexamethasone, and vincristine. Mechanistically, LDM triggered MM cells apoptosis by increasing the levels of cleaved poly ADP-ribose polymerase (PARP) and caspase-3/7. In addition, activation of p38 mitogen-activated protein kinase (MAPK) and c-Jun NH2-terminal kinase (JNK) was a critical mediator in LDM-induced cell death. Inhibition of the expression of p38 MAPK and JNK by pharmacological inhibitors reversed the LDM-induced apoptosis through decreasing the level of cleaved PARP and caspase-3/7. Interestingly, phosphorylation of extracellular signal-related kinase was increased by LDM; conversely, MEK inhibitor synergistically enhanced LDM-induced cytotoxicity and apoptosis in MM cells. The results demonstrated that LDM suppresses MM cell growth through the activation of p38 MAPK and JNK, with the potential to be developed as a chemotherapeutic agent for MM.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Enediynes/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Neoplasm Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Aminoglycosides/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enediynes/therapeutic use , Enzyme Activation/drug effects , Humans , Poly(ADP-ribose) Polymerases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...