Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Cancer Res ; 71(20): 6382-90, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21878529

ABSTRACT

Synthetic oligodeoxynucleotides expressing CpG motifs (CpG-ODN) are a Toll-like receptor 9 (TLR9) agonist that can enhance the antitumor activity of DNA-damaging chemotherapy and radiation therapy in preclinical mouse models. We hypothesized that the success of these combinations is related to the ability of CpG-ODN to modulate genes involved in DNA repair. We conducted an in silico analysis of genes implicated in DNA repair in data sets obtained from murine colon carcinoma cells in mice injected intratumorally with CpG-ODN and from splenocytes in mice treated intraperitoneally with CpG-ODN. CpG-ODN treatment caused downregulation of DNA repair genes in tumors. Microarray analyses of human IGROV-1 ovarian carcinoma xenografts in mice treated intraperitoneally with CpG-ODN confirmed in silico findings. When combined with the DNA-damaging drug cisplatin, CpG-ODN significantly increased the life span of mice compared with individual treatments. In contrast, CpG-ODN led to an upregulation of genes involved in DNA repair in immune cells. Cisplatin-treated patients with ovarian carcinoma as well as anthracycline-treated patients with breast cancer who are classified as "CpG-like" for the level of expression of CpG-ODN modulated DNA repair genes have a better outcome than patients classified as "CpG-untreated-like," indicating the relevance of these genes in the tumor cell response to DNA-damaging drugs. Taken together, the findings provide evidence that the tumor microenvironment can sensitize cancer cells to DNA-damaging chemotherapy, thereby expanding the benefits of CpG-ODN therapy beyond induction of a strong immune response.


Subject(s)
Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , DNA Repair/drug effects , Oligodeoxyribonucleotides/pharmacology , Ovarian Neoplasms/drug therapy , Spleen/drug effects , Toll-Like Receptor 9/agonists , Animals , Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Carcinoma/genetics , Cell Line, Tumor , Cisplatin/therapeutic use , Colonic Neoplasms/genetics , DNA Repair/genetics , Down-Regulation/drug effects , Female , Humans , Mice , Ovarian Neoplasms/genetics , Spleen/immunology , Treatment Outcome , Up-Regulation/drug effects
2.
Bioorg Med Chem ; 19(18): 5291-9, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21880496

ABSTRACT

Bis-2,3-heteroarylmaleimides and polyheterocondensed imides joined through nitrogen atoms of the N,N'-bis(ethyl)-1,3-propanediamine linker were prepared from substituted maleic anhydrides and symmetrical diamines in good to satisfactory yields and short reaction times using microwave heating. The novel molecules were shown to inhibit proliferation of human tumor cells (NCI-H460 lung carcinoma) and rat aortic smooth muscle cells (SMCs) with variable potencies. Compound 11a, the most potent one of the series, showed IC(50) values comparable to those observed for the leading molecule elinafide in both cell lines, but with a higher selectivity toward human tumor cells. Compound 11a affected G1/S phase transition of the cell cycle, showed in vitro DNA intercalating activity and in vivo antitumor activity. A thorough structural analysis of the 11a-DNA complex was also made by mean of NMR and computational techniques.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Aorta/drug effects , Imides/chemical synthesis , Imides/pharmacology , Maleimides/chemical synthesis , Maleimides/pharmacology , Muscle, Smooth, Vascular/drug effects , Animals , Antineoplastic Agents/chemistry , Aorta/cytology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Imides/chemistry , Maleimides/chemistry , Models, Molecular , Molecular Structure , Muscle, Smooth, Vascular/cytology , Rats , Structure-Activity Relationship
3.
Cancer Lett ; 296(2): 206-15, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20435407

ABSTRACT

There is indication that tumor growth is sustained by subpopulation of cells with stem-like features but little is known on their genomic characterization and their genetic stability. We report a detailed molecular cytogenetic characterization using Spectral Karyotyping and fluorescent in situ hybridization of parental serum-cultured adherent cells and their sphere-growing stem-like counterpart before and after differentiation from six cell lines established from solid tumors. Our findings indicate increased cytogenetic complexity in sphere-growing stem-like and their differentiated adherent cells compared to parental adherent component suggesting the existence within cell lines of heterogeneous and genetically unstable subpopulations of cells endowed with stem-like features.


Subject(s)
Cell Line, Tumor/pathology , Stem Cells/pathology , Antigens, CD/analysis , Biomarkers, Tumor/analysis , Cell Adhesion , Cell Culture Techniques/methods , Cell Differentiation , Cell Division , Chromosome Aberrations , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Karyotyping/methods , Neoplasms/genetics
4.
J Invest Dermatol ; 130(7): 1877-86, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20376064

ABSTRACT

Melanospheres, the melanoma cells that grow as nonadherent colonies and that show in vitro self-renewing capacity and multipotency, were selected from melanoma specimens or from melanoma cell lines. Melanospheres were highly tumorigenic, and intradermal injections in severe combined immunodeficient (SCID) mice of as few as 100 cells generated tumors that maintained tumorigenic potential into subsequent recipients. Primary and serially transplanted xenografts recapitulated the phenotypic features of the original melanoma of the patient. Melanoma cells cultured in the presence of fetal calf serum (FCS) were also tumorigenic in SCID mice, although with lower efficiency; these xenografts showed a homogeneous phenotype for the expression of melanoma-associated markers, Melan-A/Mart-1, HMB45, and MITF, and contained cells with features of fully differentiated cells. Melanospheres were heterogeneous for the expression of stem cell markers and showed a significantly enhanced expression of the Nanog and Oct3/4 transcription factors when compared with adherent melanoma cells. No direct and unique correlation between any of the examined stem cell markers and in vivo tumorigenicity was found. Taken together, our data provide further evidence on the heterogeneous nature of human melanomas and show that melanospheres and their corresponding tumors, which are generated in vivo in immunocompromised mice, represent a model to investigate melanoma biology.


Subject(s)
Disease Models, Animal , Melanoma/secondary , Mice, SCID , Neoplasm Transplantation/methods , Skin Neoplasms/pathology , Animals , Biomarkers, Tumor/metabolism , Blood Proteins/pharmacology , CD146 Antigen/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Homeodomain Proteins/metabolism , Humans , Immunophenotyping , In Vitro Techniques , Lymphatic Metastasis , Melanoma/physiopathology , Mice , Nanog Homeobox Protein , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/metabolism , Skin Neoplasms/physiopathology , Spheroids, Cellular , Transplantation, Heterologous
5.
J Immunother ; 33(1): 8-15, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19952960

ABSTRACT

Tumor cell growth, even in advanced stages of ovarian cancer, is nearly always restricted to the peritoneal cavity; therefore, repeated intraperitoneal injections of oligodeoxynucleotides containing dinucleotides with unmethylated CpG motifs (CpG-ODN) recruiting and activating innate effector cells throughout the abdominal cavity to the tumor site might control tumor cell growth and ascites formation. After a single CpG-ODN treatment, in IGROV-1 ovarian tumor ascites-bearing athymic mice, the number of tumor cells declined rapidly and markedly, and ascites volumes declined shortly after treatment (5 h), increasing thereafter at a slower rate than in controls. When administered every 7 days for 4 weeks, CpG-ODN had only a marginal effect on survival time, whereas administration 5 days/wk for 3 or 4 weeks led to a significantly increased survival time as compared with controls (P<0.005) and completely controlled ascites growth without apparent toxicity, although a disorganization of lymphoid organs was observed. Bio-plex assay of cytokine levels in peritoneal fluid of ascites-bearing mice after CpG-ODN treatment revealed an increase in interleukin (IL)-6, IL-10, IL-12, and interferon-gamma at 24 hours, which returned to control mice levels at 48 to 96 hours, whereas the high levels of angiogenic factors remained unchanged. Depletion of natural killer or monocytes/macrophages only slightly influenced the CpG-ODN-induced reduction of ascites tumor cells, indicating that the antitumor activity might not be related to a specific cell/cytokine but rather to the repertoire of cells and cytokines accumulated in the peritoneal cavity. Thus, our data suggest a relevant role for repeated activation of cells and cytokines of innate immunity in the therapy of ovarian cancer patients with malignant ascites.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Ascites/drug therapy , Oligodeoxyribonucleotides/administration & dosage , Ovarian Neoplasms/drug therapy , Animals , Ascites/etiology , Drug Administration Schedule , Female , Humans , Injections, Intraperitoneal , Mice , Mice, Nude , Oligodeoxyribonucleotides/immunology , Ovarian Neoplasms/complications , Ovarian Neoplasms/immunology , Xenograft Model Antitumor Assays
6.
Proc Natl Acad Sci U S A ; 106(38): 16281-6, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19805294

ABSTRACT

The identification of lung tumor-initiating cells and associated markers may be useful for optimization of therapeutic approaches and for predictive and prognostic information in lung cancer patients. CD133, a surface glycoprotein linked to organ-specific stem cells, was described as a marker of cancer-initiating cells in different tumor types. Here, we report that a CD133+, epithelial-specific antigen-positive (CD133+ESA+) population is increased in primary nonsmall cell lung cancer (NSCLC) compared with normal lung tissue and has higher tumorigenic potential in SCID mice and expression of genes involved in stemness, adhesion, motility, and drug efflux than the CD133(-) counterpart. Cisplatin treatment of lung cancer cells in vitro resulted in enrichment of CD133+ fraction both after acute cytotoxic exposure and in cells with stable cisplatin-resistant phenotype. Subpopulations of CD133+ABCG2+ and CD133+CXCR4+ cells were spared by in vivo cisplatin treatment of lung tumor xenografts established from primary tumors. A tendency toward shorter progression-free survival was observed in CD133+ NSCLC patients treated with platinum-containing regimens. Our results indicate that chemoresistant populations with highly tumorigenic and stem-like features are present in lung tumors. The molecular features of these cells may provide the rationale for more specific therapeutic targeting and the definition of predictive factors in clinical management of this lethal disease.


Subject(s)
Antigens, CD/metabolism , Cisplatin/pharmacology , Glycoproteins/metabolism , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Peptides/metabolism , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Female , Flow Cytometry , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, CXCR4/metabolism , Survival Analysis , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Int J Cancer ; 124(9): 2179-85, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19123479

ABSTRACT

N(6)-isopentenyladenosine (i(6)A), a member of the cytokinin family of plant hormones, has potent in vitro antitumour activity in different types of human epithelial cancer cell lines. Gene expression profile analysis of i(6)A-treated cells revealed induction of genes (e.g., PPP1R15A, DNAJB9, DDIT3, and HBP1) involved in the negative regulation of cell cycle progression and reportedly up-regulated during cell cycle arrest in stress conditions. Of 6 i(6)A analogues synthesized, only the 1 with a saturated double bond of the isopentenyl side chain had in vitro antitumour activity, although weaker than that of i(6)A, suggesting that i(6)A biological activity is highly linked to its structure. In vivo analysis of i(6)A and the active analogue revealed no significant inhibition of cancer cell growth in mice by either reagent. Thus, although i(6)A may inhibit cell proliferation by regulating the cell cycle, further studies are needed to identify active analogues potentially useful in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Isopentenyladenosine/chemistry , Neoplasms/drug therapy , Neoplasms/genetics , Pharmacogenetics , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Gene Expression Profiling , Humans , Isopentenyladenosine/chemical synthesis , Isopentenyladenosine/pharmacology , Mice , Mice, Nude , Molecular Structure , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Stem Cell Assay
8.
Clin Cancer Res ; 14(17): 5512-8, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18765543

ABSTRACT

PURPOSE: Oligodeoxynucleotides containing unmethylated CpG motifs (CpG-ODN) are potent activators of innate and adaptive immunity. Recognition of CpG-ODN is mediated by Toll-like receptor 9 expressed by immune cells, endothelial and epithelial cells, and fibroblasts. We examined the antitumor effect of CpG-ODN and the role of administration route on human ovarian cancers growing in the peritoneal cavity of nude mice. EXPERIMENTAL DESIGN: Mice implanted i.p. with human ovarian carcinoma cells were treated i.p., s.c., or i.v. and assessed for survival and tumor-free incidence. Peritoneal washings were analyzed for keratinocyte chemokine production and for functional and phenotypic profiles as indicators of the cell types involved in mediating the antitumor effects. RESULTS: IGROV-1-bearing mice treated i.p. survived significantly longer than those treated i.v. or s.c. (P=0.0005), and nearly half of them (8 of 17) were tumor-free by the end of the experiment, a rate never achieved using a variety of chemotherapeutic drugs. High rates of tumor-free mice were observed in three other ovarian tumor xenografts treated i.p. Compared with peritoneal washings of mice treated s.c. or i.v., those from mice treated i.p. showed the highest level of serum and tissue keratinocyte chemokine, the highest number of natural killer cells and neutrophils, and the highest antiproliferative activity in vitro. CONCLUSIONS: The superior antitumor effect obtained by locoregional administration of CpG-ODN in i.p. tumor-bearing mice with a limited adaptive immune response points to the importance of innate effector cells amplification at the site of tumor growth and suggests the promise of i.p. CpG-ODN in clinical trials for ovarian cancer.


Subject(s)
Injections, Intraperitoneal , Oligodeoxyribonucleotides/therapeutic use , Ovarian Neoplasms/therapy , Adjuvants, Immunologic/therapeutic use , Animals , Drug Administration Routes , Female , Humans , Immunotherapy , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/immunology , Transplantation, Heterologous
9.
Mol Cancer Ther ; 7(7): 2051-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645015

ABSTRACT

ST1968 is a novel hydrophilic camptothecin (CPT) derivative of the 7-oxyiminomethyl series. Because ST1968 retained ability to form remarkably stable cleavable complexes, this study was done to investigate its preclinical profile of antitumor activity in a large panel of human tumor models, including irinotecan-resistant tumors. Although less potent than SN38 in vitro, i.v. administered ST1968 caused a marked tumor inhibition, superior to that of irinotecan, in most tested models. ST1968 exhibited an impressive activity against several tumors including models of ovarian and colon carcinoma in which a high rate of cures was observed. In the most responsive tumors, complete and persistent tumor regressions were achieved even with low suboptimal doses. Even tumors derived from intrinsically resistant cells exhibited a significant responsiveness. Histologic analysis of treated tumors supports a contribution of both proapoptotic and antiangiogenic effects to ST1968 antitumor efficacy. A study done in yeast cells transformed with CPT-resistant mutant forms of topoisomerase I documented that, in contrast to other tested CPT, ST1968 was active against yeasts expressing the mutant K720E enzyme. Based on its outstanding efficacy superior to that of irinotecan and of its good therapeutic index, ST1968 has been selected for clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Animals , Camptothecin/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type I/metabolism , Female , Humans , Mice , Mice, Nude , Microbial Viability/drug effects , Mutant Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Topotecan/pharmacology , Xenograft Model Antitumor Assays
10.
J Immunother ; 31(6): 537-44, 2008.
Article in English | MEDLINE | ID: mdl-18528301

ABSTRACT

The monoclonal antibody trastuzumab binds to the extracellular domain of HER-2/neu and induces clinical responses in breast tumors with HER-2 gene amplification and/or protein overexpression. Its role in other tumor types remains to be investigated. We evaluated the antitumor efficacy of trastuzumab in vitro and in nude mice implanted orthotopically with cells of 3 human pancreatic tumor lines expressing only low levels of HER-2/neu, as determined by flow cytometry. Although none of the 3 cell lines showed growth inhibition when cultured directly with trastuzumab, 2 of them, GER and PaCa3, were sensitive to lysis in antibody-dependent cellular cytotoxicity assay. This pattern of response was recapitulated in tumor-bearing mice repeatedly treated with trastuzumab, in which survival was significantly prolonged as compared with controls (P=0.03 for GER and 0.0008 for PaCa3). Incidence of metastases was also reduced, especially in liver. These preclinical results indicate that trastuzumab can exert an antitumor effect against orthotopic human pancreatic cancer xenografts with low-level HER-2/neu expression and that this effect correlates with the in vitro antibody-dependent cellular cytotoxicity susceptibility, suggesting a different role for HER-2/neu in the therapy of tumor types other than breast cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Pancreatic Neoplasms/drug therapy , Receptor, ErbB-2/analysis , Animals , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Trastuzumab , Xenograft Model Antitumor Assays
11.
Cancer Biol Ther ; 7(4): 596-601, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18364568

ABSTRACT

Gimatecan, a potent lipophilic camptothecin, is active by oral route, which allows a comparison of different treatment schedules. The present study was designed based on previous evidence of improved antitumor efficacy of the combination of topotecan with immunostimulatory CpG-ODN. Two regimens of gimatecan, i.e., a low-dose/protracted treatment (metronomic) and a high-dose/intermittent treatment, were compared in the GER orthotopic human pancreatic tumor model in nude mice. Metronomic gimatecan (0.25 mg/kg per os, daily) significantly increased mice survival time over control mice (154 T/C%, p < 0.05), in contrast to intermittent treatment-gimatecan (1.5 mg/kg per os, q4d) or intravenous gemcitabine (125 and 128 T/C%, respectively, p > 0.1). Metronomic gimatecan combined to CpG-ODN (20 microg/mouse intraperitoneal, weekly) was well tolerated and achieved a strong antitumor effect, with a T/C% of 188 on survival time (P < 0.001 versus control mice), significantly superior to those of the single agent-treated mice (p < 0.05 versus gimatecan- or CpG-ODN-treated mice). Cellular studies indicated that TRAIL could increase the apoptotic response to gimatecan in GER tumor cells, and TRAIL was released in the peritoneal washings of CpG-ODN-treated mice. In conclusion, the combination of metronomic gimatecan with CpG-ODN was effective and tolerated, and might represent a preclinical basis for the design of clinical studies, even considering the ability of CpG-ODN to stimulate TRAIL release and the high level of TRAIL-receptors expressed in pancreatic tumor cells.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Camptothecin/analogs & derivatives , Oligodeoxyribonucleotides/therapeutic use , Pancreatic Neoplasms/drug therapy , Animals , Apoptosis , Camptothecin/therapeutic use , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Topotecan/therapeutic use , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem ; 16(5): 2431-8, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18077173

ABSTRACT

Synthesis of 10 pyrroloiminoquinone derivatives is presented. The strategy is based around the elaboration of a common intermediate by reaction with primary amines. All the compounds obtained have been subjected to antiproliferative activity with three different cell lines (NCI-H460, HeLa, and HL-60). The capacity of 4 selected compounds to affect the enzymatic activity of the nuclear enzyme DNA topoisomerase II and to form the typical DNA fragmentation which occurs in the apoptotic process is discussed here.


Subject(s)
Pyrroloiminoquinones/chemical synthesis , Pyrroloiminoquinones/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/genetics , DNA Topoisomerases, Type II/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Pyrroloiminoquinones/chemistry , Structure-Activity Relationship , Topoisomerase II Inhibitors
13.
J Pharmacol Exp Ther ; 324(1): 15-22, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17909082

ABSTRACT

On the basis of the evidence that vacuolar H(+)-ATPase is implicated in the development of the metastatic phenotype, we have explored the possibility to target the enzyme function in an attempt to control the metastatic behavior of tumor cells. In this study, we used an indole derivative, NiK-12192 [4-(5,6-dichloro-1H-indol-2-yl)-3-ethoxy-N-(2,2,6,6-tetramethyl-piperidin-4-yl)-benzamide], recently identified as an effective inhibitor of vacuolar H(+)-ATPase, as a potential antimetastatic agent in the treatment of NSCLC H460 xenograft, which is able to induce lung metastases in mice. Oral administration of NiK-12192 caused a significant inhibition of formation of spontaneous metastases. In contrast, the drug exhibited a negligible effect on the development of artificial metastases (i.e., after i.v. injection of tumor cells), thus supporting that the drug affects the early events of the metastatic process (e.g., migration and invasion). Cellular effects are consistent with this interpretation. In conclusion, the available results show for the first time that a vacuolar H(+)-ATPase inhibitor is effective in modulation of the metastatic behavior of a lung carcinoma, supporting its potential therapeutic interest as a novel treatment approach.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Indoles/therapeutic use , Lung Neoplasms/drug therapy , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Indoles/pharmacology , Integrin alphaVbeta3/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Topotecan/therapeutic use , Vacuolar Proton-Translocating ATPases/metabolism , Wound Healing/drug effects , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem ; 16(4): 1691-701, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18061459

ABSTRACT

A series of 2,3-heteroarylmaleimides 9 and polyheterocondensed imides 12 were prepared in good yields and short reaction time using a very efficient procedure consisting in the condensation of the corresponding anhydrides and N,N-diethylethylenediamine and microwave heating. The antiproliferative activity of the novel molecules was tested against human tumor cells (NCI-H460 lung carcinoma) and rat aortic smooth muscle cells (SMCs). The IC50 values for the novel molecules ranged from 0.08 to 13.9 microM in SMCs, and from 0.84 to 9 microM in the tumor cell line. The activity profile for compounds 9 and 12 is comparable to that obtained for amonafide in NCI-H460, except for fused imides 12b,i which proved to be about 10-fold more potent. Whereas, in rat SMCs, only the compound 12b was shown to be 10-fold more potent than amonafide. Instead 12c is equipotent to amonafide. These results suggest that the extended pi-system and the kind of heteroatom are essential in the binding with the molecular target.


Subject(s)
Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Imides/pharmacology , Maleimides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Aorta , Cell Line, Tumor , Cells, Cultured , Humans , Imides/chemical synthesis , Inhibitory Concentration 50 , Maleimides/chemical synthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Rats , Structure-Activity Relationship
15.
Biochem Pharmacol ; 73(5): 656-64, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17169332

ABSTRACT

In previous studies, we have documented the potential therapeutic advantages of camptothecin analogs modified at the 7-position, i.e., 7-oxyiminomethyl derivatives. The present study was performed to explore the therapeutic potential of novel hydrophilic derivatives of this series. With one exception (ST1976), the tested camptothecins exhibited a reduced antiproliferative activity and all compounds retained ability to stabilize the topoisomerase I-mediated cleavable complex. The two analogs (ST1976 and ST1968) characterized by the presence of a free amino group in the side chain also exhibited the formation of persistent cleavable complexes. The most potent compound, ST1976 (7-(4-aminobenzyl)oxyiminomethylcamptothecin), was selected for evaluation of its preclinical profile of antitumor activity in a large panel of human tumor xenografts. As expected on the basis of the introduction of a hydrophilic substituent, the novel camptothecin was a substrate for BCRP. However, in spite of an apparent recognition by BCRP, ST1976 was effective following oral administration. The antitumor activity was evaluated using various schedules and routes of administration (i.v. and p.o.). ST1976 exhibited a remarkable activity in all tested tumors and was effective in a number of tumors which are resistant to irinotecan. The biological and pharmacological profile of ST1976 supports the therapeutic potential of camptothecins containing hydrophilic substituents at the 7-position. On the basis of its excellent activity in preclinical models, ST1976 is a promising candidate for clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Animals , Antineoplastic Agents/administration & dosage , Camptothecin/administration & dosage , Camptothecin/pharmacology , Cell Line, Tumor , DNA Cleavage/drug effects , DNA Topoisomerases, Type I/metabolism , Drug Administration Schedule , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Female , Humans , Irinotecan , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy
16.
J Nat Prod ; 69(12): 1796-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17190463

ABSTRACT

A reinvestigation of the fruiting bodies of the mushroom Leucopaxillus gentianeus, allowed the isolation of two minor cucurbitane triterpenes, namely, cucurbitacin D (5) and the new metabolite 16-deoxycucurbitacin B (6). The latter compound lacks an oxygenated substituent at C-16, an unprecedented structural feature among congeners of cucurbitacin B. The cucurbitanes present in the fruiting bodies were compared with those extracted from mycelia grown on the modified Melin-Norkans (MMN) culture medium. Cucurbitacins B (1) and D (5), as well as leucopaxillones A (3) and B (4), were isolated from both sources; in contrast, 16-deoxycucurbitacin B (6) and a mixture of fatty acid esters of cucurbitacin B (2) were absent in the mycelia. A new triterpene, 18-deoxyleucopaxillone A (7), was isolated from the mycelia, but was not detected in the fruiting bodies. The antiproliferative activity of the isolated triterpenes was determined against the NCI-H460 human tumor cell line, in comparison with the antitumor compound topotecan, a well-known topoisomerase I inhibitor.


Subject(s)
Agaricales/chemistry , Antineoplastic Agents/isolation & purification , Topoisomerase I Inhibitors , Triterpenes/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Fruiting Bodies, Fungal/chemistry , Humans , Molecular Structure , Mycelium/chemistry , Rome , Topotecan/pharmacology , Triterpenes/chemistry , Triterpenes/pharmacology
17.
Cancer Res ; 66(22): 10976-82, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108136

ABSTRACT

A series of water-soluble camptothecins obtained by linking a spermidine moiety to the 21-position of the open form through an amidic bond have been tested for their biochemical and biological activities. Growth inhibition assay on the human non-small cell lung cancer carcinoma NCI-H460 cell line revealed that the camptothecin analogues were less potent than topotecan and SN38 after 1 hour of treatment. The potency increased after 72 hours of exposure, being similar to that of reference camptothecins. The analysis of topoisomerase I-mediated DNA cleavage using the purified enzyme indicated that the novel camptothecin analogues retained ability to poison topoisomerase I and displayed the same cleavage pattern of SN38. Persistence of the DNA cleavage was comparable with that of SN38. Stabilization of the cleavable complex was not the result of hydrolysis of the N-C bond between polyamine and the drug because no free camptothecin was recovered at the end of DNA cleavage in presence of IDN5174, the analogue selected for detailed studies. IDN5174 exhibited an antitumor activity comparable with that of topotecan and irinotecan against NCI-H460 tumor xenograft. The pharmacokinetics in mice showed a favorable disposition in tumor tissue with low amount of camptothecin detectable in plasma and tumor (around 5-10%), thus supporting the efficacy of intact IDN5174. In conclusion, we found that IDN5174 maintained the biological and antitumor properties, in spite of lack of the closed E ring. The available results support the interpretation that the polyamine linked at the 21-position may allow a favorable drug interaction in the ternary complex.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Humans , Lactones/pharmacokinetics , Lactones/pharmacology , Mice , Spermidine/analogs & derivatives , Xenograft Model Antitumor Assays
18.
Mol Cancer Ther ; 5(9): 2388-97, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985073

ABSTRACT

Hepatocyte growth factor (HGF) and its tyrosine kinase receptor Met play a pivotal role in the tumor metastatic phenotype and represent attractive therapeutic targets. We investigated the biochemical and biological effects of the tyrosine kinase inhibitor RPI-1 on the human lung cancer cell lines H460 and N592, which express constitutively active Met. RPI-1-treated cells showed down-regulation of Met activation and expression, inhibition of HGF/Met-dependent downstream signaling involving AKT, signal transducers and activators of transcription 3 and paxillin, as well as a reduced expression of the proangiogenic factors vascular endothelial growth factor and basic fibroblast growth factor. Cell growth in soft agar of H460 cells was strongly reduced in the presence of the drug. Furthermore, RPI-1 inhibited both spontaneous and HGF-induced motility/invasiveness of both H460 and human endothelial cells. Targeting of Met signaling by alternative methods (Met small interfering RNA and anti-phosphorylated Met antibody intracellular transfer) produced comparable biochemical and biological effects. Using the spontaneously metastasizing lung carcinoma xenograft H460, daily oral treatment with well-tolerated doses of RPI-1 produced a significant reduction of spontaneous lung metastases (-75%; P < 0.001, compared with control mice). In addition, a significant inhibition of angiogenesis in primary s.c. tumors of treated mice was observed, possibly contributing to limit the development of metastases. The results provide preclinical evidence in support of Met targeting pharmacologic approach as a new option for the control of tumor metastatic dissemination.


Subject(s)
Indoles/pharmacology , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/blood supply , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/pathology , Cell Adhesion/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/pharmacology , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/metabolism , Xenograft Model Antitumor Assays
19.
Biochem Pharmacol ; 72(4): 405-14, 2006 Aug 14.
Article in English | MEDLINE | ID: mdl-16756963

ABSTRACT

Recent evidence indicates that the success of molecular targeted therapies may depend on the identification of drug targets which are essential for the survival of subsets of tumors. RET oncogenes that have been implicated in the development of thyroid carcinomas are emerging as potential therapeutic targets. In the present study, we investigated the efficacy and the cellular bases of antitumor activity of the indolinone Ret tyrosine kinase inhibitor RPI-1 against large established s.c. TT tumor xenograft, a human medullary thyroid carcinoma (MTC) harboring oncogenic MEN-2A-type RET mutation. Oral treatment with RPI-1 caused growth arrest or regression in 81% treated tumors. Following treatment suspension, tumor inhibition was maintained (51%, P<0.05, 100 days) and cures were achieved in 2/11 mice. In treated tumors, Ret was tyrosine dephosphorylated. Moreover, compared to control tumors, a significant increase in apoptotic cells (210%, P<0.0001), loss of cellularity (47%, P<0.0001) and reduction of microvessel density (36%, P<0.0005) were detected. In vivo effects of RPI-1 were reflected in activation of BAD, cleavage of caspases, apoptotic DNA fragmentation and inhibition of VEGF production observed in in vitro RPI-1-treated TT cells. These findings thus indicate that RPI-1 antitumor effect on the MTC was characterized by apoptosis induction and angiogenesis inhibition. The results, consistent with a dependence on RET oncogene activation for maintenance and survival of MEN2A-type MTC, provide further preclinical rationale for a pharmacological RET-targeted intervention in thyroid cancer.


Subject(s)
Apoptosis/drug effects , Carcinoma, Medullary/drug therapy , Indoles/pharmacology , Neovascularization, Pathologic/prevention & control , Thyroid Neoplasms/drug therapy , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Medullary/pathology , Cell Line, Tumor , Female , Flow Cytometry , Humans , Indoles/administration & dosage , Indoles/therapeutic use , Mice , Mice, Nude , Mutation , NIH 3T3 Cells , Phosphorylation/drug effects , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
20.
J Pharmacol Exp Ther ; 318(3): 939-46, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16714402

ABSTRACT

The vacuolar-H(+)-ATPase, functionally expressed in cell membranes, is known to play a relevant role in intracellular pH regulatory mechanisms, because it is implicated in pumping protons into the extracellular environment or in sequestrating excess protons into acidic vacuolar compartments. Because tumor cells exist in a hypoxic microenvironment and produce acidic metabolites, this regulatory mechanism is recognized as a protective function. This study was designed to investigate the effect of NiK-12192 [4-(5,6-dichloro-1H-indol-2-yl)-3-ethoxy-N-(2,2,6,6-tetramethyl-piperidin-4-yl)-benzamide], an indole derivative identified as an effective inhibitor of vacuolar-H(+)-ATPase, on the cytotoxic activity of two camptothecins, i.e., topotecan and SN-38 (7-ethyl-10-hydroxycamptothecin, the active metabolite of irinotecan). The cellular studies performed in two pairs of human colon carcinoma cell lines, i.e., LoVo and LoVo/DX (overexpressing P-glycoprotein) and HT29 and HT29/Mit (overexpressing breast cancer resistant protein), indicated an enhancement of the antiproliferative effect of camptothecins by concomitant exposure to subtoxic concentrations of NiK-12192. Studies of subcellular distribution indicated that whereas topotecan alone localized mainly in mitochondria and endoplasmic compartment, the simultaneous presence of NiK-12192 caused a cytoplasmic redistribution. In HT29/Mit cells, NiK-12192 reverted the pattern of acidification induced by topotecan. The potentiation of topotecan efficacy by NiK-12192 was documented by an increased efficacy of the combination in both the HT29 tumor xenografts, being more evident in the topotecan-resistant HT29/Mit tumor. In conclusion, the vacuolar-H(+)-ATPase inhibitor NiK-12192 was able to potentiate the cytotoxic/antitumor effects of camptothecins, either in in vitro or in in vivo systems. Such findings support a potential interest for the use of vacuolar-H(+)-ATPase inhibitors in combination therapy to improve camptothecin efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Camptothecin/analogs & derivatives , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Topotecan/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/analysis , Animals , Camptothecin/pharmacology , Female , HT29 Cells , Humans , Irinotecan , Mice , Neoplasm Proteins/analysis , Neoplasms, Experimental/drug therapy , Staining and Labeling , Topotecan/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...