Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Anticancer Agents Med Chem ; 17(13): 1741-1755, 2017.
Article in English | MEDLINE | ID: mdl-28530540

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzimidazoles/chemistry , Benzothiazoles/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , Drug Stability , Humans , Rats
3.
Anticancer Agents Med Chem ; 17(7): 1003-1013, 2017.
Article in English | MEDLINE | ID: mdl-27924733

ABSTRACT

AIMS: A series of 1,4-dihydropyridine based compounds bearing benzylpyridinium moiety have been designed and evaluated for in vitro anticancer activity against glioblastoma U87MG, lung cancer A549 and colorectal adenocarcinoma Caco-2 cell lines using the MTT assay. METHOD: Among these compounds, 7b, 7d, 7e, and 7f exhibited potent anticancer activity against the cell lines tested. The cytotoxicity of the synthesized derivatives was compared to standard drugs (carboplatin, gemcitabine, and daunorubicin). RESULT: Thus, synthesized 1,4-dihydropyridines can be considered as the encouraging molecules for further drug development as anticancer agents.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dihydropyridines/chemistry , Dihydropyridines/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Dihydropyridines/chemical synthesis , Drug Screening Assays, Antitumor , Humans , Neoplasms/drug therapy , Structure-Activity Relationship
4.
Oncotarget ; 7(25): 38078-38090, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27191263

ABSTRACT

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.


Subject(s)
Autophagy/drug effects , Quinolines/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Neoplasms/metabolism , Transfection
5.
Oncotarget ; 6(28): 25770-83, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26208482

ABSTRACT

Alkylating agents are frequently used as first-line chemotherapeutics for various newly diagnosed cancers. Disruption of genome integrity by such agents can lead to cell lethality if DNA lesions are not removed. Several DNA repair mechanisms participate in the recovery of mono- or bi-functional DNA alkylation. Thus, DNA repair capacity is correlated with the therapeutic response. Here, we assessed the function of novel water-soluble N-mustard BO-1055 (ureidomustin) in DNA damage response and repair mechanisms. As expected, BO-1055 induces ATM and ATR-mediated DNA damage response cascades, including downstream Chk1/Chk2 phosphorylation, S/G2 cell-cycle arrest, and cell death. Further investigation revealed that cell survival sensitivity to BO-1055 is comparable to that of mitomycin C. Both compounds require nucleotide excision repair and homologous recombination, but not non-homologous end-joining, to repair conventional cross-linking DNA damage. Interestingly and unlike mitomycin C and melphalan, MGMT activity was also observed in BO-1055 damage repair systems, which reflects the occurrence of O-alkyl DNA lesions. Combined treatment with ATM/ATR kinase inhibitors significantly increases BO-1055 sensitivity. Our study pinpoints that BO-1055 can be used for treating tumors that with deficient NER, HR, and MGMT DNA repair genes, or for synergistic therapy in tumors that DNA damage response have been suppressed.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Damage , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , Nitrogen Mustard Compounds/pharmacology , Phenylurea Compounds/pharmacology , Recombinational DNA Repair , Tumor Suppressor Proteins/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , CHO Cells , Cell Death/drug effects , Cricetulus , Dose-Response Relationship, Drug , Drug Synergism , HEK293 Cells , Humans , MCF-7 Cells , Melphalan/pharmacology , Mitomycin/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA Interference , S Phase Cell Cycle Checkpoints/drug effects , Signal Transduction/drug effects , Time Factors , Transfection
6.
Eur J Med Chem ; 83: 695-708, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25014640

ABSTRACT

We synthesized a series of phenyl N-mustard-4-anilinoquinoline conjugates to study their antitumorigenic effects. These agents were prepared by the condensation of 4-[N,N-bis(2-chloroethyl)amino]phenyl isocyanate with 6-amino-4-methylamino or 4-anilinoquinolines. The structure-activity relationship (SAR) studies revealed that the C2-methylquinoline derivatives (18a-o) were generally more cytotoxic than the C2-phenylquinoline conjugates (23a-d) in inhibiting the cell growth of various human tumor cell lines in vitro. However, the methylamino or aniline substituents at C4 of quinoline did not influence the cytotoxic effects. The title conjugates were capable of inducing DNA cross-linking and promoting cell-cycle arrest at the G2/M phase. This study demonstrates that phenyl N-mustard-4-anilinoquinoline conjugates are generally more potent than phenyl N-mustard-4-anilinoquinazoline conjugates against the cell growth of various tumor cell-lines.


Subject(s)
Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/pharmacology , DNA/metabolism , Quinolines/chemistry , Urea/chemistry , Antineoplastic Agents, Alkylating/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Solubility , Structure-Activity Relationship
7.
Transl Oncol ; 7(2): 256-266.e5, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24913674

ABSTRACT

The synergistic targeting of DNA damage and DNA repair is a promising strategy for the development of new chemotherapeutic agents for human lung cancer. The DNA interstrand cross-linking agent BO-1509, a derivative of 3a-aza-cyclopenta[α]indene, was synthesized and combined with the phosphoinositide 3-kinase (PI3K) inhibitor LY294002 to treat human lung cancer cells. Our results showed that the BO-1509 and LY294002 combination synergistically killed lung cancer cells in culture and also suppressed the growth of lung cancer xenografts in mice, including those derived from gefitinib-resistant cells. We also found that LY294002 suppressed the induction of several DNA repair proteins by BO-1509 and inhibited the nuclear translocation of Rad51. On the basis of the results of the γH2AX foci formation assays, LY294002 apparently inhibited the repair of DNA damage that was induced by BO-1509. According to the complete blood profile, biochemical enzyme analysis, and histopathologic analysis of major organs, no apparent toxicity was observed in mice treated with BO-1509 alone or in combination with LY294002. Our results suggest that the combination of a DNA cross-linking agent with a PI3K inhibitor is a feasible strategy for the treatment of patients with lung cancer.

8.
Toxicol Appl Pharmacol ; 278(3): 249-58, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24823293

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma/drug therapy , Drug Synergism , Enzyme Inhibitors/pharmacology , Protein Kinases/metabolism , Antineoplastic Agents, Alkylating/agonists , Autophagy-Related Protein 7 , Carbamates/agonists , Carbamates/pharmacology , Carcinoma/enzymology , Carcinoma/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Checkpoint Kinase 1 , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class III Phosphatidylinositol 3-Kinases/metabolism , Cross-Linking Reagents/pharmacology , Female , Gene Silencing , HeLa Cells , Heterocyclic Compounds, 3-Ring/agonists , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Indenes/agonists , Indenes/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Processing, Post-Translational/drug effects , S Phase/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism
9.
Eur J Med Chem ; 76: 155-69, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24583355

ABSTRACT

A series of new, water-soluble phenyl N-mustard-benzenealkylamide conjugates containing hydrophilic ω-dialkylaminoalkylamide or ω-cyclic aminoalkylamide moieties were synthesized via a bioisostere approach. These compounds have a broad spectrum of antitumor activity against a panel of human tumor cell lines. Of these derivatives, compound 18b effectively suppressed the growth of colon cancer (HCT-116), prostate cancer (PC3), and lung cancer (H460) xenografts. The growth of HCT-116 xenografts was almost completely suppressed when co-treated with compound 18b and 5-fluorouracil. Furthermore, compound 18b can induce DNA cross-linking and cell-cycle arrest at the G2/M phase. Early preclinical studies, including pharmacokinetics in rats, inhibition of the hERG, and 14 days of acute intravenous injection toxicity, suggest that compound 18b is a promising candidate for further preclinical studies.


Subject(s)
Antineoplastic Agents/chemistry , Mustard Compounds/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , In Situ Nick-End Labeling , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Nude , Mustard Compounds/chemical synthesis , Mustard Compounds/pharmacology , Rats , Spectrometry, Mass, Electrospray Ionization
10.
Eur J Med Chem ; 69: 609-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24095754

ABSTRACT

Bifunctional DNA cross-linking agents are widely used as chemotherapeutic agents in clinics. The advance in the development of these agents as potential antitumor agents has generated various types of bis(hydroxymethyl)pyrrole analogs. In order to develop highly effective anticancer agents, it is necessary to understand the chemophysical properties, structure-activity relationships, therapeutic potency, toxicity/safety, and pharmacokinetics of these DNA cross-linking agents. This review presents an overview of the recent advances in developing various types of bis(hydroxymethyl)pyrrole analogs with potential antitumor activity to provide more information for future drug design and strategies for combination chemotherapy. The rational drug design, chemical syntheses, antitumor activity, mechanism of action, and development of combined chemotherapy regimens, including a DNA repair inhibitor, are discussed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cross-Linking Reagents/pharmacology , DNA Repair/drug effects , DNA/drug effects , Pyrroles/pharmacology , Pyrrolidines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/chemical synthesis , Antineoplastic Combined Chemotherapy Protocols/chemistry , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/chemistry , DNA/chemistry , Humans , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry
11.
J Med Chem ; 56(4): 1544-63, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23360284

ABSTRACT

A series of bis(hydroxymethyl)indolizino[6,7-b]indoles and their bis(alkylcarbamates) were synthesized for antitumor studies. These agents were designed as hybrid molecules of ß-carboline (topoisomerase inhibition moiety) and bis(hydroxymethyl)pyrrole (DNA cross-linking moiety). The preliminary antitumor studies indicated that these agents exhibited significant cytotoxicity against a variety of human tumor cells in vitro. Treatment of human breast carcinoma MX-1 xenograft-bearing nude mice with compounds 18b and 28c achieved more than 99% tumor remission. We also observed that 18a displayed potent therapeutic efficacy against human lung adenocarcinoma A549 and colon cancer HT-29 xenografts. These results revealed that compound 18a was more potent than irinotecan against HT-29 cells and was as potent as irinotecan against A549 cells in xenograft models. Furthermore, we demonstrated that these derivatives possess multiple modes of action, such as induction of DNA cross-linking, inhibition of topoisomerase I and II, and cell-cycle arrest at the S-phase.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cross-Linking Reagents/chemical synthesis , DNA/metabolism , Indoles/chemical synthesis , Indolizines/chemical synthesis , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Indoles/pharmacology , Indolizines/chemistry , Indolizines/pharmacology , Intercalating Agents/chemical synthesis , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Rats , S Phase Cell Cycle Checkpoints/drug effects , Structure-Activity Relationship , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Transplantation, Heterologous
12.
Article in English | MEDLINE | ID: mdl-23353940

ABSTRACT

Ureidomustin hydrochloride (BO-1055) was designed as a water-soluble nitrogen-mustard, which exhibited potent anticancer activity and was selected as a candidate for preclinical studies. However, up to date, there is rarely an easy and economic method to quantize ureidomustin in the biological samples. The aim of this study is to develop a simple yet valid quantization method to tackle this challenge. Here we present a combined high-performance liquid chromatography with photodiode array (HPLC-PDA) method in quantizing the ureidomustin in the plasma and various organs of Sprague-Dawley rats. The method was validated in terms of precision, accuracy, and extraction recovery. Furthermore, the established method was applied to study pharmacokinetics of ureidomustin in the rat's plasma and verified via a liquid chromatography tandem mass spectrometry (LC-MS/MS) method. Calibration curves of the plasma and organ samples were falling at the range between 0.5-50µg/mL and 0.1-50µg/mL (r(2)≥0.999 and CV≤±15%), respectively. The limits of detection (LOD) were 0.1µg/mL for plasma samples and 0.05µg/mL for organ samples, while the detection limits of quantification (LOQ) were 0.5µg/mL for plasma samples and 0.1µg/mL for organ samples. The average recovery of ureidomustin was about 83%. These results demonstrated a linear pharmacokinetic pattern at dosages of 10 and 30mg/kg. The pharmacokinetic data revealed that ureidomustin was best fitted to a two-compartment model with a rapid distribution phase and a slow elimination phase. Besides, after a short intravenous administration time at the dose of 10mg/kg, ureidomustin was found to be quickly distributed to all organs in rats, accumulated mainly in the kidney, and only a limited amount was detected in the brain.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Nitrogen Mustard Compounds/pharmacokinetics , Phenylurea Compounds/pharmacokinetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Limit of Detection , Male , Nitrogen Mustard Compounds/blood , Nitrogen Mustard Compounds/chemistry , Phenylurea Compounds/blood , Phenylurea Compounds/chemistry , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Tandem Mass Spectrometry/methods , Tissue Distribution
13.
Eur J Med Chem ; 53: 28-40, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22507893

ABSTRACT

A series of novel 2,3-bis(hydroxymethyl)benzo[d]pyrrolo[2,1-b]thiazoles and their bis(alkylcarbamate) derivatives were synthesized starting from benzothiazole via reaction with dimethyl acetylenedicarboxylate (DMAD)/tetra-fluoro boric acid, catalytic hydrogenation, and alkylcarbamoylation. The anti-proliferative activity of these agents against human leukemia and various solid tumor cell growth in vitro was studied. The structure-activity relationship studies revealed that the bis(alkylcarbamates) derivatives are generally more cytotoxic than the corresponding bis(hydroxymethyl) congeners in inhibiting human lymphoblastic leukemia CCRF-CEM and various human solid tumor cell growth in culture. These agents have no cross-resistance to taxol or vinblastine. Studies on the therapeutic effect against human breast carcinoma MX-1 xenograft showed that complete tumor remission (CR) were achieved by treating with C1-4'-F- or C1-4'-Cl-Ph-bis(i-propylcarbamates) derivatives (19b and 19c, respectively) and more than 99% tumor suppression by the corresponding bis(ethylcarbamates) 18b and 18c at the maximal tolerated dose. Alkaline agarose gel shifting assay revealed that the newly synthesized compounds are able to induce DNA interstrand cross-linking. The present studies generated a series of new potent DNA interstrand cross-linking agents, which have potential for further antitumor drug development.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , DNA/chemistry , Humans , Inhibitory Concentration 50 , Male , Mice , Thiazoles/chemistry , Xenograft Model Antitumor Assays
14.
Int J Cancer ; 130(6): 1440-50, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-21500194

ABSTRACT

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.


Subject(s)
Alkylating Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , DNA/drug effects , Mouth Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , CHO Cells , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle Checkpoints/drug effects , Cell Growth Processes/drug effects , Cell Line , Cell Line, Tumor , Cricetinae , DNA Damage , Fibroblasts/drug effects , G1 Phase/drug effects , Humans , KB Cells , Mice , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , S Phase/drug effects
15.
ChemMedChem ; 6(6): 1119-29, 2011 Jun 06.
Article in English | MEDLINE | ID: mdl-21542133

ABSTRACT

A series of 2-substituted quinolin-4-yl-benzenesulfonate derivatives were synthesized for the purpose of evaluating antiproliferative activity. Structure-activity relationships of the newly synthesized compounds against human lymphoblastic leukemia and various solid tumor cell growths in culture are discussed. Of these derivatives, 2-phenyl-6-pyrrolidinyl-4-quinoline sulfonate analogues 10 f, 10 g, and 10 k, and 4'-nitrophenyl sulfonate 10 m exhibit superior cytotoxicity over other sulfonates. The antiproliferative activities of these compounds correlate well with their abilities to induce mitotic arrest and apoptosis. Mechanistic studies indicate that they target the vinblastine binding site of tubulin and inhibit cellular tubulin polymerization. Hence, these compounds induce the formation of aberrant mitotic spindles and mitotic arrest, resulting in intensive apoptosis. The tested compounds were shown to be poor substrates for membrane multidrug resistance transporters. The present studies suggest that these newly synthesized compounds are promising tubulin polymerization inhibitors and are worthy of further investigation as antitumor agents.


Subject(s)
4-Quinolones/pharmacology , Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , 4-Quinolones/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzenesulfonates/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Mitosis/drug effects , Neoplasms/drug therapy , Tubulin Modulators/chemistry
16.
Bioorg Med Chem ; 19(6): 1987-98, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21356592

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/chemical synthesis , Quinazolines/chemistry , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Cell Line, Tumor , Drug Design , Humans , Mice , Mice, Nude , Mustard Plant/chemistry , Neoplasms/drug therapy , Structure-Activity Relationship , Urea/chemistry , Xenograft Model Antitumor Assays
17.
Bioorg Med Chem ; 19(1): 275-86, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21144756

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/pharmacology , Isoquinolines/chemical synthesis , Isoquinolines/pharmacology , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Xenograft Model Antitumor Assays
18.
Bioorg Med Chem ; 19(1): 471-85, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21106377

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Design , Mustard Compounds/chemical synthesis , Mustard Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Limit of Detection , Male , Mice , Mice, Nude , Mustard Compounds/chemistry , Rats , Rats, Sprague-Dawley , Solubility , Water , Xenograft Model Antitumor Assays
19.
Neoplasia ; 12(5): 376-87, 2010 May.
Article in English | MEDLINE | ID: mdl-20454509

ABSTRACT

Drug resistance is a crucial factor in the failure of cancer chemotherapy. In this study, we explored the effect of combining alkylating agents and arsenic trioxide (ATO) on the suppression of tumor cells with inherited or acquired resistance to therapeutic agents. Our results showed that combining ATO and a synthetic derivative of 3a-aza-cyclopenta[a]indenes (BO-1012), a bifunctional alkylating agent causing DNA interstrand cross-links, was more effective in killing human cancer cell lines (H460, H1299, and PC3) than combining ATO and melphalan or thiotepa. We further demonstrated that the combination treatment of H460 cells with BO-1012 and ATO resulted in severe G(2)/M arrest and apoptosis. In a xenograft mouse model, the combination treatment with BO-1012 and ATO synergistically reduced tumor volumes in nude mice inoculated with H460 cells. Similarly, the combination of BO-1012 and ATO effectively reduced the growth of cisplatin-resistant NTUB1/P human bladder carcinoma cells. Furthermore, the repair of BO-1012-induced DNA interstrand cross-links was significantly inhibited by ATO, and consequently, gammaH2AX was remarkably increased and formed nuclear foci in H460 cells treated with this drug combination. In addition, Rad51 was activated by translocating and forming foci in nuclei on treatment with BO-1012, whereas its activation was significantly suppressed by ATO. We further revealed that ATO might mediate through the suppression of AKT activity to inactivate Rad51. Taken together, the present study reveals that a combination of bifunctional alkylating agents and ATO may be a rational strategy for treating cancers with inherited or acquired drug resistance.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arsenicals/administration & dosage , Carbamates/administration & dosage , Cell Proliferation/drug effects , Heterocyclic Compounds, 3-Ring/administration & dosage , Neoplasms, Experimental/drug therapy , Oxides/administration & dosage , Signal Transduction/drug effects , Animals , Apoptosis , Arsenic Trioxide , Blotting, Western , Cell Line, Tumor , Cell Separation , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
20.
Bioorg Med Chem ; 18(6): 2285-2299, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20181487

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA/chemistry , DNA/drug effects , Hydrazines/chemistry , Neoplasms, Experimental/drug therapy , Quinolines/pharmacology , Urea/chemistry , Animals , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/chemistry , Cell Proliferation/drug effects , DNA/metabolism , Drug Screening Assays, Antitumor , Humans , Male , Mice , Mice, Nude , Molecular Structure , Neoplasm Transplantation , Quinolines/chemical synthesis , Quinolines/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...