Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 403
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 21(11)2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32545380

ABSTRACT

Glioblastoma (GBM) present with an abundant and aberrant tumor neo-vasculature. While rapid growth of solid tumors depends on the initiation of tumor angiogenesis, GBM also progress by infiltrative growth and vascular co-option. The angiogenic factor apelin (APLN) and its receptor (APLNR) are upregulated in GBM patient samples as compared to normal brain tissue. Here, we studied the role of apelin/APLNR signaling in GBM angiogenesis and growth. By functional analysis of apelin in orthotopic GBM mouse models, we found that apelin/APLNR signaling is required for in vivo tumor angiogenesis. Knockdown of tumor cell-derived APLN massively reduced the tumor vasculature. Additional loss of the apelin signal in endothelial tip cells using the APLN-knockout (KO) mouse led to a further reduction of GBM angiogenesis. Direct infusion of the bioactive peptide apelin-13 rescued the vascular loss-of-function phenotype specifically. In addition, APLN depletion massively reduced angiogenesis-dependent tumor growth. Consequently, survival of GBM-bearing mice was significantly increased when APLN expression was missing in the brain tumor microenvironment. Thus, we suggest that targeting vascular apelin may serve as an alternative strategy for anti-angiogenesis in GBM.


Subject(s)
Apelin/metabolism , Brain Neoplasms/blood supply , Glioblastoma/blood supply , Neovascularization, Pathologic/pathology , Animals , Apelin/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Magnetic Resonance Imaging , Mice, Knockout , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/mortality , Neovascularization, Pathologic/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
2.
Planta Med ; 83(1-02): 151-157, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27352383

ABSTRACT

Eight ethanolic extracts of indigenous Taiwanese plants of the genus Alpinia were tested for tumor cytotoxicity against AGS, Hep G2, HeLa, KB, and HL-60 cells. Among the 50 % and 95 % EtOH extracts of eight Alpinia species, the cytotoxic effects of Alpinia intermedia leaves were the strongest. When the leaf extract of A. intermedia was partitioned using n-hexane and aqueous solvents, the n-hexane layer showed a greater cytotoxic effect and could prolong the survival time of P-388D1 tumor-bearing CDF1 mice. Two new labdane diterpene derivatives, intermedin A (1) and intermedin B (2), and coronarin E (3) were isolated from the n-hexane layer of A. intermedia. Intermedin A induced apoptosis in HL-60 cells at 30 µg/mL and significantly prolonged the survival time of P-388D1 tumor-bearing CDF1 mice by 48.7 % at 20 mg/kg of body weight. We suggest that intermedin A is a major compound of A. intermedia and has a cytotoxic effect on HL-60 and P-388D1 cells.


Subject(s)
Alpinia/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Diterpenes/pharmacology , Lignans/pharmacology , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Cell Line, Tumor , Diterpenes/chemistry , Diterpenes/isolation & purification , Ethanol , Humans , Lignans/chemistry , Lignans/isolation & purification , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/mortality , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacology
3.
Gastroenterology ; 144(5): 1098-106, 2013 May.
Article in English | MEDLINE | ID: mdl-23333712

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDA) is an aggressive tumor, and patients typically present with late-stage disease; rates of 5-year survival after pancreaticoduodenectomy are low. Antibodies against α-enolase (ENO1), a glycolytic enzyme, are detected in more than 60% of patients with PDA, and ENO1-specific T cells inhibit the growth of human pancreatic xenograft tumors in mice. We investigated whether an ENO1 DNA vaccine elicits antitumor immune responses and prolongs survival of mice that spontaneously develop autochthonous, lethal pancreatic carcinomas. METHODS: We injected and electroporated a plasmid encoding ENO1 (or a control plasmid) into Kras(G12D)/Cre (KC) mice and Kras(G12D)/Trp53(R172H)/Cre (KPC) mice at 4 weeks of age (when pancreatic intraepithelial lesions are histologically evident). Antitumor humoral and cellular responses were analyzed by histology, immunohistochemistry, enzyme-linked immunosorbent assays, flow cytometry, and enzyme-linked immunosorbent spot and cytotoxicity assays. Survival was analyzed by Kaplan-Meier analysis. RESULTS: The ENO1 vaccine induced antibody and a cellular response and increased survival times by a median of 138 days in KC mice and 42 days in KPC mice compared with mice given the control vector. On histologic analysis, the vaccine appeared to slow tumor progression. The vaccinated mice had increased serum levels of anti-ENO1 immunoglobulin G, which bound the surface of carcinoma cells and induced complement-dependent cytotoxicity. ENO1 vaccination reduced numbers of myeloid-derived suppressor cells and T-regulatory cells and increased T-helper 1 and 17 responses. CONCLUSIONS: In a genetic model of pancreatic carcinoma, vaccination with ENO1 DNA elicits humoral and cellular immune responses against tumors, delays tumor progression, and significantly extends survival. This vaccination strategy might be developed as a neoadjuvant therapy for patients with PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Immunity, Cellular/immunology , Neoplasms, Experimental/drug therapy , Pancreatic Neoplasms/drug therapy , Phosphopyruvate Hydratase/immunology , Vaccination/methods , Vaccines, DNA/pharmacology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Mutant Strains , Neoplasms, Experimental/genetics , Neoplasms, Experimental/mortality , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Survival Rate
4.
Cancer Immunol Immunother ; 63(6): 615-26, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24682539

ABSTRACT

Immune responses wane during aging, posing challenges to the potential effectiveness of cancer immunotherapies. We previously demonstrated that in the context of a promising immunotherapeutic, OX40 agonist (αOX40), older animals exhibited impaired anti-tumor immune responses and diminished CD4 T cell effector differentiation. In this study, we hypothesized that tumor immune responses could be maintained during aging through caloric restriction (CR) or dietary supplementation with resveratrol (RES), a CR mimetic. Mice were placed on either a calorically restricted diet or a RES-formulated diet starting between 4 and 6 months of age and continued until mice reached 12 months of age. Tumor immune responses were assessed after challenging with either sarcoma or breast tumor cells followed by αOX40 treatment. Our results show that CR, but not RES, maintained OX40-mediated anti-tumor immunity. In addition, CR fully sustained antigen-specific CD4 T cell priming in aged hosts (12 months old), whereas tumor-specific CD8 T cell priming was not fully maintained compared to young reference animals (2 months old). Thus, CR appears to maintain immunological fitness of the CD4 T cell priming environment during aging, which is critical for optimal OX40-mediated responses.


Subject(s)
Aging/immunology , Anticarcinogenic Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , Caloric Restriction , Neoplasms, Experimental/immunology , Receptors, OX40/agonists , Stilbenes/pharmacology , Adoptive Transfer , Animals , Blotting, Western , Female , Flow Cytometry , Humans , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms, Experimental/mortality , Neoplasms, Experimental/therapy , Resveratrol , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology
5.
Mol Carcinog ; 53(8): 610-24, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23401136

ABSTRACT

The KAI1/CD82 tetraspanin is a widely expressed cell surface molecule thought to organize diverse cellular signaling processes. KAI1/CD82 suppresses metastasis but not tumorigenicity, establishing it as one of a class of metastasis suppressor genes. In order to further assess its functions, we have characterized the phenotypic properties of Kai1/Cd82 deleted mice, including viability, fertility, lymphocyte composition, blood chemistry and tissue histopathology, and of their wild-type and heterozygote littermates. Interestingly, Kai1/Cd82(-/-) showed no obvious genotype associated defects in any of these processes and displayed no genotype associated histopathologic abnormalities after 12 or 18 months of life. Expression profiles of non-immortal, wild-type and Kai1/Cd82(-/-) mouse embryo fibroblast (MEFs) indicated distinct sex-specific and genotype-specific profiles. These data identify 191 and 1,271 differentially expressed transcripts (by twofold at P < 0.01) based on Kai1/CD82 genotype status in female and male MEFs, respectively. Differentially expressed genes in male MEFs were surprisingly enriched for cell division related processes, suggesting that Kai1/Cd82 may functionally affect these processes. This suggests that Kai/Cd82 has an unappreciated role in the early establishment of proliferation and division when challenged with a new environment that might play a role in adaptability to new metastatic sites.


Subject(s)
Cell Proliferation , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Kangai-1 Protein/physiology , Neoplasms, Experimental/mortality , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Northern , Blotting, Western , Cells, Cultured , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
6.
Carcinogenesis ; 34(10): 2361-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23803690

ABSTRACT

Gemcitabine has limited clinical benefits in pancreatic ductal adenocarcinoma. The solvent-based traditional taxanes docetaxel and paclitaxel have not shown clinical results superior to gemcitabine. Nab-paclitaxel, a water-soluble albumin-bound paclitaxel, may carry superior distribution properties into the tumor microenvironment and has shown efficacy in multiple tumor types. We evaluated nab-paclitaxel effects compared with gemcitabine or docetaxel. For pancreatic ductal adenocarcinoma cells AsPC-1, BxPC-3, MIA PaCa-2 and Panc-1, gemcitabine IC50 ranged from 494nM to 23.9 µM; docetaxel IC50 range was from 5 to 34nM; nab-paclitaxel IC50 range was from 243nM to 4.9 µM. Addition of IC25 dose of docetaxel or nab-paclitaxel decreased gemcitabine IC50. Net tumor growth inhibition after gemcitabine, docetaxel or nab-paclitaxel was 67, 31 and 72%, which corresponded with intratumoral proliferative and apoptotic indices. Tumor stromal density was decreased by nab-paclitaxel and to a lesser extent by docetaxel as measured through reduction in α-smooth muscle actin, S100A4 and collagen 1 expression. Animal survival was prolonged after nab-paclitaxel treatment (41 days, P < 0.002) compared with gemcitabine (32 days, P = 0.005), docetaxel (32 days, P = 0.005) and controls (20 days). Survival in nab-paclitaxel/gemcitabine and docetaxel/gemcitabine sequential treatment groups was not superior to nab-paclitaxel alone. Low-dose combination of gemcitabine with nab-paclitaxel or docetaxel was more effective compared with controls or gemcitabine alone but not superior to regular dose nab-paclitaxel alone. Combination treatment of gemcitabine+nab-paclitaxel or gemcitabine+docetaxel increased gemcitabine concentration in plasma and tumor. The superior antitumor activity of nab-paclitaxel provides a strong rationale for considering nab-paclitaxel as first-line monotherapy in pancreatic ductal adenocarcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Neoplasms, Experimental/drug therapy , Paclitaxel/pharmacology , Pancreatic Neoplasms/drug therapy , Taxoids/pharmacology , Albumin-Bound Paclitaxel , Albumins/administration & dosage , Albumins/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Disease Models, Animal , Docetaxel , Female , Humans , Inhibitory Concentration 50 , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Paclitaxel/administration & dosage , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Stathmin/metabolism , Stromal Cells/drug effects , Taxoids/administration & dosage , Tubulin/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
7.
Mol Ther ; 20(2): 432-42, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21934655

ABSTRACT

In vivo dendritic cells (DC) targeting is an attractive approach with potential advantages in vaccine efficacy, cost, and availability. Identification of molecular adjuvants to in vivo "modulate " DC to coordinately render improved Th1 and CD8 T cell immunity, and attenuated deleterious Treg effects, is a critical challenge. Here, we report that in vivo genetic targeting of the active transcription factor XBP1s to DC (XBP1s/DC) potentiated vaccine-induced prophylactic and therapeutic antitumor immunity in multiple tumor models. This immunization strategy is based on a genetic vaccine encoding both cytomegalovirus (CMV)-driven vaccine Aghsp70 and DC-specific CD11c-driven XBP1s. The novel targeted vaccine induced durable Th1 and CD8 T cell responses to poorly immunogenic self/tumor antigen (Ag) and attenuated tumor-associated Treg suppressive function. Bone marrow (BM)-derived DC genetically modified to simultaneously overexpress XBP1s and express Aghsp70 upregulated CD40, CD70, CD86, interleukin (IL)-15, IL-15Rα, and CCR7 expression, and increased IL-6, IL-12, and tumor necrosis factor (TNF)-α production in vitro. XBP1s/DC elevated functional DEC205(+)CD8α(+)DC in the draining lymph nodes (DLN). The data suggest a novel role for XBP1s in modulating DC to potentiate tumor vaccine efficacy via overcoming two major obstacles to tumor vaccines (i.e., T cell hyporesponsiveness against poorly immunologic self/tumor Ag and tumor-associated Treg-mediated suppression) and improving DEC205(+)CD8α(+)DC.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Gene Targeting , Neoplasms, Experimental/immunology , Transcription Factors/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cell Survival/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Female , Gene Expression , Gene Order , Interferon-gamma/biosynthesis , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/mortality , Regulatory Factor X Transcription Factors , Survival Analysis , T-Lymphocytes, Regulatory , TRPC Cation Channels/immunology , Th1 Cells/immunology , Transcription Factors/metabolism , Vaccines, DNA/genetics , Vaccines, DNA/immunology , X-Box Binding Protein 1
8.
J Vasc Interv Radiol ; 23(12): 1685-91, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23177115

ABSTRACT

PURPOSE: To characterize tumor growth of N1S1 cells implanted into the liver of Sprague-Dawley rats to determine if this model could be used for survival studies. These results were compared with tumor growth after implantation with McA-RH7777 cells. MATERIALS AND METHODS: N1S1 or McA-RH7777 cells were implanted into the liver of Sprague-Dawley rats (n = 20 and n = 12, respectively) using ultrasound (US) guidance, and tumor growth was followed by using US. Serum profiles of 19 cytokines were compared in naive versus tumor-bearing rats. RESULTS: Both types of tumors were visible on US 1 week after tumor implantation, but the mean tumor volume of N1S1 tumors was larger compared to McA-RH7777 tumors (231 mm(3) vs 82.3 mm(3), respectively). Tumor volumes in both groups continued to increase, reaching means of 289 mm(3) and 160 mm(3) in N1S1 and McA-RH7777 groups, respectively, 2 weeks after tumor implantation. By week 3, tumor volumes had decreased considerably, and six tumors (50%) in the McA-RH7777 had spontaneously regressed, versus two (10%) in the N1S1 group. Tumor volumes continued to decrease over the following 3 weeks, and complete tumor regression of all tumors was seen 5 weeks and 6 weeks after tumor implantation in the McA-RH7777 and N1S1 groups, respectively. In an N1S1-implanted rat, multiple cytokines that have been shown to correlate with the ability of the tumor to survive in a hostile environment were increased by as much as 50%, whereas the average increase in cytokine levels was 90%. These findings suggest that the net cytokine environment favors an antitumor immune response. A similar trend was observed in a rat with a McA-RH7777 tumor, and the increase in cytokine levels was considerably more pronounced, with an average increase of 320%. CONCLUSIONS: The model of N1S1 cell implantation in the liver of Sprague-Dawley rats is not ideal for survival studies and should only be used with great caution in short-term studies that involve cancer therapies.


Subject(s)
Disease Models, Animal , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/mortality , Neoplasm Regression, Spontaneous , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/mortality , Animals , Cell Line, Tumor , Humans , Rats , Rats, Sprague-Dawley , Survival Analysis , Survival Rate , Transplantation, Isogeneic , Ultrasonography
9.
Pediatr Blood Cancer ; 58(2): 191-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21337679

ABSTRACT

BACKGROUND: AZD8055 is a small molecule ATP-competitive inhibitor of the serine/threonine kinase mTOR that regulates cap-dependent translation through the mTORC1 complex and Akt activation through the mTORC2 complex. Procedures AZD8055 was tested against the PPTP in vitro panel at concentrations ranging from 1.0 nM to 10 µM and against the PPTP in vivo panels at a dose of 20 mg/kg administered orally daily x 7 for 4 weeks. RESULTS: In vitro the median relative IC(50) for AZD8055 against the PPTP cell lines was 24.7 nM. Relative I/O values >0% (consistent with a cytostatic effect) were observed in 8 cell lines and 15 cell lines showed Relative I/O values ranging from -4.7 to -92.2% (consistent with varying degrees of cytotoxic activity). In vivo AZD8055 induced significant differences in EFS distribution compared to controls in 23 of 36 (64%) evaluable solid tumor xenografts, and 1 of 6 evaluable ALL xenografts. Intermediate activity for the time to event activity measure (EFS T/C >2) was observed in 5 of 32 (16%) solid tumor xenografts evaluable. The best response was stable disease. PD2 (progressive disease with growth delay) was observed in 20 of 36 (55.6%) evaluable solid tumor xenografts. AZD8055 significantly inhibited 4E-BP1, S6, and Akt phosphorylation following day 1 and day 4 dosing, but suppression of mTORC1 or mTORC2 signaling did not predict tumor sensitivity. CONCLUSIONS: AZD8055 demonstrated broad activity in vitro, but at the dose and schedule studied demonstrated limited activity in vivo against the PPTP solid tumor and ALL panels.


Subject(s)
Cell Proliferation/drug effects , Morpholines/therapeutic use , Neoplasms, Experimental/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Blotting, Western , Child , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/mortality , Survival Rate , Tumor Cells, Cultured
10.
Pediatr Blood Cancer ; 58(2): 200-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21548007

ABSTRACT

BACKGROUND: Genz-644282 is a novel non-camptothecin topoisomerase I poison that is in clinical development. PROCEDURES: Genz-644282 was tested against the PPTP in vitro panel (0.1 nM to 1 µM), and in vivo using three times per week × 2 schedule repeated at day 21 at its maximum tolerated dose (MTD) of 4 mg/kg. Subsequently Genz-644282 was tested at 4, 3, 2, and 1 mg/kg in 3 models to assess the dose-response relationship. mRNA gene signatures predictive for Genz-644282 response in vitro were applied to select 15 tumor models that were evaluated prospectively. RESULTS: In vitro, Genz-644282 demonstrated potent cytotoxic activity with a median IC(50) of 1.2 nM (range 0.2-21.9 nM). In vivo, Genz-644282 at its MTD (4 mg/kg) induced maintained complete responses (MCR) in 6/6 evaluable solid tumor models. At 2 mg/kg Genz-644282 induced CR or MCR in 3/3 tumor models relatively insensitive to topotecan, but there were no objective responses at 1 mg/kg. Further testing at 2 mg/kg showed that Genz-644282 induced objective regressions in 7 of 17 (41%) models. There was a significant correlation between predictive response scores based on Affymetrix U133Plus2 baseline tumor expression profiles and the observed in vivo responses to Genz-644282. CONCLUSIONS: Genz-644282 was highly active within a narrow dose range (2-4 mg/kg), typical of other topoisomerase I poisons. As with other topoisomerase I poisons, how accurately these data will translate to clinical activity will depend upon the drug exposures that can be achieved in children treated with this agent.


Subject(s)
Cell Proliferation/drug effects , DNA Topoisomerases, Type I/chemistry , Naphthyridines/therapeutic use , Neoplasms, Experimental/drug therapy , Topoisomerase I Inhibitors/therapeutic use , Animals , Child , DNA Topoisomerases, Type I/metabolism , Disease-Free Survival , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/mortality , Survival Rate , Topotecan/therapeutic use , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
J Immunol ; 185(2): 892-901, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20543102

ABSTRACT

The optimization of anticancer therapeutic vaccines can lead to better efficacy but also to stronger adverse effects. In a mouse model of antitumor vaccination using a long peptide (LP), which included MHC class I- and II-restricted male (H-Y) epitopes, we observed unexpected mortality. Mice with an increased frequency of anti-H-Y CD4 T cells were primed with LP+CpG and boosted 10 d later. Within hours of boost, they displayed shock-like signs with high mortality. Serum cytokine levels were high. TNF-alpha secreted by the CD4 T cells was identified as the key effector molecule. Priming with a short peptide (SP), which included the MHC class II-restricted epitope, was a more efficient primer than LP, but did not lead to mortality when used as boost. The high mortality induced by LP compared with SP was probably related to its specific ability to be presented by B cells. Finally, targeting the LP sequence to dendritic cells allowed tumor protection without side effects. Our data: 1) confirm that the immune system can be very dangerous; 2) caution against the use of systemic activation of high-frequency Ag-specific T cells as induced by high doses of LP; and 3) underline the benefit of targeting Ag to dendritic cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cytokines/metabolism , Vaccines, Subunit/immunology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Dose-Response Relationship, Drug , Epitopes/immunology , Female , Major Histocompatibility Complex/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Survival Rate , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Vaccination/adverse effects , Vaccination/methods , Vaccines, Subunit/administration & dosage
12.
Eur Rev Med Pharmacol Sci ; 16(13): 1753-64, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23208959

ABSTRACT

OBJECTIVE: The two scaffold Quinazoline analogues (Compound 21, NSC: 95112/753439 and Compound 12, NSC: D-104834/ 758270) in three different concentrations were evaluated for their anti-tumor activity against Ehrlich ascites carcinoma (EAC) and two different concentration were evaluated for their anti-tumor activity against Dalton's ascites lymphoma (DLA) bearing Swiss albino mice. MATERIALS AND METHODS: The in vivo anti-tumor potency of Quinazoline bases was assessed in EAC model by measuring the increase in mean survival time of the drug treated over untreated control mice and treated standard (Gefitinib) mice. Their toxicity was assessed in vivo in normal, standard, and EAC-bearing mice by measuring the drug-induced changes in haematological parameters. The in vivo anti-tumor potency of Quinazoline bases was assessed in DLA model by measuring solid tumor volume, solid tumor weight and % inhibition of the tumor weight of the drug treated over untreated control mice and treated standard (Gefitinib) mice. RESULTS AND CONCLUSIONS: Among the two quinazoline bases studied, 3-(2-chloro benzylideneamine)-2-(furan-2-yl) quinazoline-4(3h)-one (Compound 21) at an optimal dose of 20 mg/kg body weight was found to enhance the mean survival time of infected mice. Haematological parameters and mean survival time in tumor bearing mice were found to be significantly restored towards normal after treatment with Compound 21 at 20 mg/kg body weight of mice in EAC model. Tumor volume and tumor weight in tumor bearing mice were found to be significantly restored towards normal after treatment with Compound 12 at 20 mg/kg body weight of mice in DLA model. Compound 21 at a prime dose of 20 mg has shown promising anticancer activity in vivo against EAC and DLA models when compared to standard drug with minimum toxic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms, Experimental/drug therapy , Quinazolines/pharmacology , Animals , Body Weight/drug effects , Cell Line, Tumor , Female , Mice , Neoplasms, Experimental/mortality
13.
J Clin Invest ; 118(9): 3170-80, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18704196

ABSTRACT

Iatrogenic tumor cell implantation within surgical wounds can compromise curative cancer surgery. Adhesion of cancer cells, in particular colon cancer cells, is stimulated by exposure to increased extracellular pressure through a cytoskeleton-dependent signaling mechanism requiring FAK, Src, Akt, and paxillin. Mechanical stimuli during tumor resection may therefore negatively impact patient outcome. We hypothesized that perioperative administration of colchicine, which prevents microtubule polymerization, could disrupt pressure-stimulated tumor cell adhesion to surgical wounds and enhance tumor-free survival. Ex vivo treatment of Co26 and Co51 colon cancer cells with colchicine inhibited pressure-stimulated cell adhesion to murine surgical wounds and blocked pressure-induced FAK and Akt phosphorylation. Surgical wound contamination with pressure-activated Co26 and Co51 cells significantly reduced tumor-free survival compared with contamination with tumor cells under ambient pressure. Mice treated with pressure-activated Co26 and Co51 cells from tumors preoperatively treated with colchicine in vivo displayed reduced surgical site implantation and significantly increased tumor-free survival compared with mice exposed to pressure-activated cells from tumors not pretreated with colchicine. Our data suggest that pressure activation of malignant cells promotes tumor development and impairs tumor-free survival and that perioperative colchicine administration or similar interventions may inhibit this effect.


Subject(s)
Colchicine/pharmacology , Neoplasms, Experimental/drug therapy , Wound Healing/drug effects , Animals , Cell Line, Tumor , Disease-Free Survival , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/mortality , Paxillin/metabolism , Phosphorylation , Pressure , Proto-Oncogene Proteins c-akt/metabolism , Tubulin Modulators/pharmacology , src-Family Kinases/metabolism
14.
Immunol Invest ; 40(2): 130-49, 2011.
Article in English | MEDLINE | ID: mdl-20923331

ABSTRACT

As observed in most molluscan hemocyanins, high-mannose type glycans were identified in hemocyanins from Rapana venosa (RvH), Helix lucorum (HlH) and keyhole limpet (Megatura crenulata). In addition, a glycan with a branching structure containing xylose, fucose and terminal methyl hexose was identified in ß-HlH. We have examined the immuno-adjuvant properties of hemocyanins, their derivatives and conjugates associated with the cell mediated immunity in experimental tumor-bearing animals with ascites tumor of Guerin. After immunization of the animals with the experimental vaccine preparations, the highest values of splenic lymphocytes were observed in groups immunized with the conjugates RvH-TAg, ß-HlH-TAg and KLH-TAg (42.3%; 40.8% and 40.58%, respectively) than with the native hemocyanins (36.5%; 35.1% and 32.4%, respectively). The immunization of rats with the hemocyanins ß-HlH, RvH and KLH and their conjugates, prolonged the median survival time of tumor-bearing animals compared with non-immunized animals (39, 33, 31 and 7 days, respectively). Both hemocyanins ß-HlH and RvH activate the immune system of the experimental animals and therefore could be a good alternative for KLH. For this reason they could be included into the composition of non-specific anti-tumor vaccines to enhance their effectiveness.


Subject(s)
Antineoplastic Agents/pharmacology , Hemocyanins/pharmacology , Mollusca/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Cancer Vaccines/immunology , Glycopeptides/analysis , Glycopeptides/chemistry , Glycopeptides/isolation & purification , Glycosylation , Immunization , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mitogens/pharmacology , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Rats , Spleen/cytology , Spleen/drug effects , Survival Analysis , Tandem Mass Spectrometry
15.
Cell Rep ; 36(8): 109537, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433039

ABSTRACT

Increased DNA replication and metastasis are hallmarks of cancer progression, while deregulated proliferation often triggers sustained replication stresses in cancer cells. How cancer cells overcome the growth stress and proceed to metastasis remains largely elusive. Proliferating cell nuclear antigen (PCNA) is an indispensable component of the DNA replication machinery. Here, we show that phosphorylation of PCNA on tyrosine 211 (pY211-PCNA) regulates DNA metabolism and tumor microenvironment. Abrogation of pY211-PCNA blocks fork processivity, resulting in biogenesis of single-stranded DNA (ssDNA) through a MRE11-dependent mechanism. The cytosolic ssDNA subsequently induces inflammatory cytokines through a cyclic GMP-AMP synthetase (cGAS)-dependent cascade, triggering an anti-tumor immunity by natural killer (NK) cells to suppress distant metastasis. Expression of pY211-PCNA is inversely correlated with cytosolic ssDNA and associated with poor survival in patients with cancer. Our results pave the way to biomarkers and therapies exploiting immune responsiveness to target metastatic cancer.


Subject(s)
Neoplasms, Experimental/immunology , Proliferating Cell Nuclear Antigen/immunology , Tumor Escape , Tumor Microenvironment/immunology , Animals , Female , Humans , MCF-7 Cells , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/mortality , Phosphorylation , Proliferating Cell Nuclear Antigen/genetics , Tumor Microenvironment/genetics , Tyrosine/genetics , Tyrosine/immunology
16.
J Exp Med ; 158(6): 2095-107, 1983 Dec 01.
Article in English | MEDLINE | ID: mdl-6644239

ABSTRACT

Injection of DBA/2, C57Bl/6, or BALB/c mice with antibody to mouse interferon alpha/beta enhanced the i.p. transplantability of six different murine tumors, as manifested by an increase in the percentage of tumor-bearing mice and a decrease in survival time. The effect was observed in mice injected with antibody to interferon raised in three sheep, a goat, and a rabbit, but not with sheep antibody to "impurities" present in the mouse interferon preparations or with normal sheep or goat globulins. The enhancement in transplantability was most marked when tumor cells had been previously passaged in vitro and were of low tumorigenicity. Analysis of some of the experimental conditions using interferon-sensitive and interferon-resistant lines of Friend erythroleukemia cells (FLC) showed that the enhancing effect was observed over a wide range of tumor cell inocula, was directly related to the amount of antibody to interferon injected and was most pronounced when antibody was administered at the time of tumor cell injection. Enhancement was also observed when FLC were injected subcutaneously (s.c.). Antibody did not act directly on the tumor cells in vitro. Although we were unable to demonstrate any biologically active interferon in mice before or after tumor cell inoculation, the results suggest that endogenous interferon is present and plays a role in inhibiting the transplantability of some murine tumors in immunocompetent mice.


Subject(s)
Interferon Type I/physiology , Neoplasms, Experimental/immunology , Animals , Antibodies/administration & dosage , Female , Graft Survival , Interferon Type I/immunology , Male , Mice , Mice, Inbred Strains , Neoplasm Transplantation , Neoplasms, Experimental/mortality , Time Factors
17.
Cancer Sci ; 101(1): 210-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19832844

ABSTRACT

c-Met is the cellular receptor for hepatocyte growth factor (HGF) and is known to be dysregulated in various types of human cancers. Activation of the HGF/c-Met pathway causes tumor progression, invasion, and metastasis. Vascular endothelial growth factor (VEGF) is also known as a key molecule in tumor progression through the induction of tumor angiogenesis. Because of their key roles in tumor progression, these pathways provide attractive targets for therapeutic intervention. We have generated a novel, orally active, small molecule compound, E7050, which inhibits both c-Met and vascular endothelial growth factor receptor (VEGFR)-2. In vitro studies indicate that E7050 potently inhibits phosphorylation of both c-Met and VEGFR-2. E7050 also potently represses the growth of both c-met amplified tumor cells and endothelial cells stimulated with either HGF or VEGF. In vivo studies using E7050 showed inhibition of the phosphorylation of c-Met and VEGFR-2 in tumors, and strong inhibition of tumor growth and tumor angiogenesis in xenograft models. Treatment of some tumor lines containing c-met amplifications with high doses of E7050 (50-200 mg/kg) induced tumor regression and disappearance. In a peritoneal dissemination model, E7050 showed an antitumor effect against peritoneal tumors as well as a significant prolongation of lifespan in treated mice. Our results indicate that E7050 is a potent inhibitor of c-Met and VEGFR-2 and has therapeutic potential for the treatment of cancer.


Subject(s)
Aminopyridines/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms, Experimental/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , Mice , Neoplasms, Experimental/mortality , Phosphorylation , Proto-Oncogene Proteins c-met/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
18.
Cancer Sci ; 101(8): 1866-74, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20579075

ABSTRACT

Pirarubicin is a derivative of doxorubicin with improved intracellular uptake and reduced cardiotoxicity. We have prepared a micellar formulation of pirarubicin using styrene-maleic acid copolymer (SMA) of mean molecular weight of 1.2 kDa, which exhibits a mean diameter of 248 nm in solution. Being a macromolecule, SMA-pirarubicin micelles exhibit excellent tumor targeting capacity due to the enhanced permeability and retention (EPR) effect. Here we report the antitumor activity of SMA-pirarubicin micelles on human colon and breast cancer cell lines in vitro, and a murine liver metastasis model in vivo. Metastatic tumor microvasculature, necrosis, apoptosis, proliferation, and survival were also investigated using immunohistochemistry for Ki-67, active caspase-3, and CD34, respectively. Drug cytotoxicity in vitro was assessed using MTT (3-[4,5-dimethyl-2-thiazolyl]-2, 5-diphenyl-2H-tetrazolium bromide) assay. In vivo, SMA-pirarubicin was administered at 100, 150, or 200 mg/kg (pirarubicin equivalent). Tumor microvasculature was also assessed using scanning electron microscopy. Styrene-maleic acid copolymer (SMA)-pirarubicin micelles were toxic against human colorectal and breast cancer cells in vitro. IC(50) was at or below 1 muM, free pirarubicin equivalent. In vivo, SMA-pirarubicin at 100 mg/kg reduced tumor volume by 80% and achieved a survival rate of 93% at 40 days after tumor inoculation. Styrene-maleic acid copolymer (SMA)-pirarubicin micelles demonstrated potent antitumor activity in this liver metastases model, contributing to prolonged survival. Histological examination of tumor nodules showed significant reduction and proliferation of tumor cells (>90%). The present results suggest that investigation of the effect of multiple dosing at later time points to further improve survival is warranted.


Subject(s)
Doxorubicin/analogs & derivatives , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms, Experimental/secondary , Maleates/administration & dosage , Neoplasms, Experimental/drug therapy , Polystyrenes/administration & dosage , Animals , Apoptosis/drug effects , Caspase 3/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/administration & dosage , Humans , Male , Mice , Mice, Inbred CBA , Micelles , Necrosis , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology
19.
Toxicol Pathol ; 38(2): 292-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20124501

ABSTRACT

It is sometimes difficult to assess the relevance of tumors that occur in treated animals in short-term studies. This report is intended to establish a general profile of tumor occurrence in young Han Wistar rats. Data were collected and evaluated from 29 rat carcinogenicity studies and from a few 2-, 4-, 13-, and 26-week studies conducted between 1995 and 2009 at Huntingdon Life Sciences, UK. The route of administration was dietary, oral gavage, or inhalation, and the analysis was confined to sporadic deaths (decedents) in carcinogenicity studies. In Han Wistar rats, the most common and earliest occurring tumor was malignant lymphoma in both sexes, the earliest being seen in the 16th and 26th week in males and females, respectively. The incidence of malignant lymphoma was slightly higher in males than in females. The second most common type of tumor was brain tumors in males and mammary tumors in females. Compared with Sprague-Dawley rats, where the most common early tumor was pituitary tumor in females, the most common early tumor in Han Wistar rats was malignant lymphoma in both sexes. These early tumor profiles are consistent with the lifetime tumor occurrence in these strains.


Subject(s)
Neoplasms, Experimental/epidemiology , Animals , Brain Neoplasms/epidemiology , Brain Neoplasms/mortality , Female , Lymphoma/epidemiology , Lymphoma/mortality , Male , Mammary Neoplasms, Animal/epidemiology , Mammary Neoplasms, Animal/mortality , Neoplasms, Experimental/mortality , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sex Factors
20.
J Enzyme Inhib Med Chem ; 25(4): 513-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20128638

ABSTRACT

A series of mononuclear Ru(II) complexes of the type [Ru(S)(2)(K)](2+), where S = 1,10-phenanthroline/2,2'-bipyridine and K = 4-OH-btsz, 4-CH(3)-btsz, 3,4-di-OCH(3)-btsz, 4-OH-binh, 4-CH(3)-binh, 3,4-di-OCH(3)-binh, were prepared and characterized by elemental analysis, FTIR, (1)H-NMR, and mass spectroscopy. The complexes displayed metal-ligand charge transfer (MLCT) transitions in the visible region. These ligands formed bidentate octahedral ruthenium complexes. The title complexes were evaluated for their in vivo anticancer activity against a transplantable murine tumor cell line, Ehrlisch's ascites carcinoma (EAC), and in vitro cytotoxic activity against human cancer cell lines Molt 4/C(8) and CEM and murine tumor cell line L1210. The ruthenium complexes showed promising biological activity especially in decreasing tumor volume and viable ascites cell counts. Treatment with these complexes prolonged the life span of mice bearing EAC tumors by 10-52%. In vitro evaluation of these ruthenium complexes revealed cytotoxic activity from 0.21 to 24 muM against Molt 4/C(8), 0.16 to 19 microM against CEM, and 0.75 to 32 microM against L1210.


Subject(s)
Antineoplastic Agents/chemical synthesis , Organometallic Compounds/chemical synthesis , Ruthenium , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Ligands , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/mortality , Organometallic Compounds/pharmacology , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL