Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 50(5): 695-704, 2020 05.
Article in English | MEDLINE | ID: mdl-31943150

ABSTRACT

CD5 is expressed on T cells and a subset of B cells (B1a). It can attenuate TCR signalling and impair CTL activation and is a therapeutic targetable tumour antigen expressed on leukemic T and B cells. However, the potential therapeutic effect of functionally blocking CD5 to increase T cell anti-tumour activity against tumours (including solid tumours) has not been explored. CD5 knockout mice show increased anti-tumour immunity: reducing CD5 on CTLs may be therapeutically beneficial to enhance the anti-tumour response. Here, we show that ex vivo administration of a function-blocking anti-CD5 MAb to primary mouse CTLs of both tumour-naïve mice and mice bearing murine 4T1 breast tumour homografts enhanced their capacity to respond to activation by treatment with anti-CD3/anti-CD28 MAbs or 4T1 tumour cell lysates. Furthermore, it enhanced TCR signalling (ERK activation) and increased markers of T cell activation, including proliferation, CD69 levels, IFN-γ production, apoptosis and Fas receptor and Fas ligand levels. Finally, CD5 function-blocking MAb treatment enhanced the capacity of CD8+ T cells to kill 4T1-mouse tumour cells in an ex vivo assay. These data support the potential of blockade of CD5 function to enhance T cell-mediated anti-tumour immunity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , CD28 Antigens/immunology , CD5 Antigens/immunology , Mammary Neoplasms, Experimental/drug therapy , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antineoplastic Agents, Immunological , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD5 Antigens/antagonists & inhibitors , CD5 Antigens/genetics , Cell Extracts/pharmacology , Cell Line, Tumor , Cell Proliferation , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/immunology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/genetics , fas Receptor/immunology
2.
J Pharmacol Exp Ther ; 364(1): 46-54, 2018 01.
Article in English | MEDLINE | ID: mdl-29061656

ABSTRACT

Although cancer cell genetic instability contributes to characteristics that mediate tumorigenicity, it also contributes to the tumor-selective toxicity of some chemotherapy drugs. This synthetic lethality can be enhanced by inhibitors of DNA repair. To exploit this potential Achilles heel, we tested the ability of a RAD51 inhibitor to potentiate the cytotoxicity of chemotherapy drugs. 2-(Benzylsulfonyl)-1-(1H-indol-3-yl)-1,2-dihydroisoquinoline (IBR2) inhibits RAD51-mediated DNA double-strand break repair but also enhances cytotoxicity of the Bcr-Abl inhibitor imatinib. The potential for synergy between IBR2 and more drugs was examined in vitro across a spectrum of cancer cell lines from various tissues. Cells were exposed to IBR2 simultaneously with inhibitors of receptor tyrosine kinases, DNA-damaging agents, or microtubule disruptors. IBR2, at concentrations that inhibited proliferation between 0% and 75%, enhanced toxicity by up to 80% of imatinib and regorafenib (targets RAF and kit); epidermal growth factor receptor inhibitors erlotinib, gefitinib, afatinib, and osimertinib; and vincristine, an inhibitor of microtubule function. However, IBR2 antagonized the action of olaparib, cisplatin, melphalan, and irinotecan. A vincristine-resistant squamous cell line was not cross resistant to imatinib, but IBR2 and another RAD51 inhibitor (B02) enhanced imatinib toxicity in this cell line, its HN-5a parent, and the colon cancer line HT-29 by up to 60% and much better than verapamil, a P-glycoprotein inhibitor (P < 0.05). Given the disparate agents the functions of which are enhanced by IBR2, the mechanisms of enhancement may be multimodal. Whether RAD51 is common to these mechanisms remains to be elucidated, but it provides the potential for selectivity to tumor cells.


Subject(s)
Cell Proliferation/drug effects , Indoles/administration & dosage , Microtubule Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Rad51 Recombinase/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Tetrahydroisoquinolines/administration & dosage , A549 Cells , Antineoplastic Agents/administration & dosage , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Drug Synergism , HEK293 Cells , HT29 Cells , Humans , K562 Cells , MCF-7 Cells , Microtubule Proteins/metabolism , Rad51 Recombinase/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
3.
Oncotarget ; 7(15): 20825-39, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26959114

ABSTRACT

Human tumor heterogeneity promotes therapeutic failure by increasing the likelihood of resistant cell subpopulations. The PARP-1 inhibitor olaparib is approved for use in BRCA-mutated ovarian cancers but BRCA2-reversion mutations lead to functional homologous recombination repair (HRR) and olaparib resistance. To overcome that resistance and expand use of PARP1 inhibition to cancers with functional HRR, we developed an antisense strategy to render the majority of tumor cells in a population BRCA2-deficient. We predicted that this strategy would render HRR-proficient tumor cells sensitive to olaparib and prevent emergence of resistance in a tumor cell population heterogeneous for HRR proficiency. We report that BRCA2 downregulation sensitized multiple human tumor cell lines (but not non-cancer human kidney cells) to olaparib and, combined with olaparib, increased aneuploidy and chromosomal translocations in human tumor cells. In a mixed HRR-proficient and HRR-deficient cell population, olaparib monotherapy allowed outgrowth of HRR-proficient cells resistant to subsequent olaparib treatment. Combined BRCA2 inhibition and olaparib treatment prevented selection of HRR-proficient cells and inhibited proliferation of the entire population. Treatment with BRCA2 siRNA and olaparib decreased ovarian xenograft growth in mice more effectively than either treatment alone. In vivo use of BRCA2 antisense oligonucleotides may be a viable option to expand clinical use of olaparib and prevent resistance.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA2 Protein/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/drug therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Animals , Apoptosis/drug effects , BRCA2 Protein/genetics , Biomarkers, Tumor/genetics , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Recombinational DNA Repair/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Tumori ; 102(1): 31-9, 2016.
Article in English | MEDLINE | ID: mdl-26429639

ABSTRACT

AIMS AND BACKGROUND: In light of the need for more selective anticancer therapy, much work has been directed at developing compounds or biological agents that target functions specific to cancer cells. To this end, numerous viruses have been engineered to exploit the dependence of cancer cells on particular anomalies that contribute to their rogue proliferative activity, such as dysfunctional p53, overactive mitogenic signaling, or a defective interferon response. The oncolytic human adenovirus dl1520 (ONYX-015) was engineered to propagate specifically in p53-deficient tumors, which comprise over half of all tumors. Based on successes in clinical trials, the full potential of dl1520 and other oncolytic viruses may be even better realized by using them in combination with conventional chemotherapy drugs. METHODS: As a model system in which to test this potential, representative cell lines from 2 common cancer types, oral squamous cell carcinoma (HN-5a) and colon adenocarcinoma (HT-29), were chosen, as well as platinum-drug-resistant variants of each. RESULTS: Following preliminary screening of virus and drug combinations, dl1520 and melphalan were found to synergistically inhibit proliferation of all the cancer cell lines. Melphalan pretreatment or cotreatment with dl1520 enhanced inhibition of proliferation by dl1520 by up to 60% and increased apoptosis by up to 25%. The tight-junction protein CAR (coxsackie and adenovirus receptor), via which adenovirus enters cells, was not upregulated by treatment with melphalan, suggesting that other mechanisms contribute to synergy. CONCLUSIONS: The synergy between melphalan and dl1520 suggests that tumor-selective cell killing by oncolytic viruses may be augmented by combining with cytotoxic drugs.


Subject(s)
Adenoviridae , Antineoplastic Agents/pharmacology , Melphalan/pharmacology , Neoplasms/drug therapy , Oncolytic Viruses , Viral Vaccines/pharmacology , Adenocarcinoma/drug therapy , Antineoplastic Agents, Alkylating/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Drug Synergism , Humans , Mouth Neoplasms/drug therapy
5.
PLoS One ; 10(11): e0143435, 2015.
Article in English | MEDLINE | ID: mdl-26579709

ABSTRACT

Indoleamine 2,3-dioxygenase-1 (IDO) is an immune regulatory enzyme expressed by most human tumors. IDO levels in tumor cells correlate with increased metastasis and poor patient outcome and IDO is linked to tumor cell resistance to immunotherapy, radiation therapy, and chemotherapy. Knowledge of tumor cell-autonomous effects of IDO, independent of its well-known role in regulating and suppressing anti-tumor immune responses, is limited. Clonal populations of A549 human lung adenocarcinoma cells stably transfected with anti-IDO shRNA or scrambled control shRNA were used to study IDO effects on drug sensitivity and resistance. IFNγ was used to induce IDO in those cells. We show, for the first time, that IDO mediates human tumor cell resistance to the candidate anticancer drugs FK866 (an NAD+ inhibitor), methoxyamine (MX, a base excision repair [BER] inhibitor) and approved anticancer drugs pemetrexed (a folate anti-metabolite) and gemcitabine (a nucleoside analogue), and combined treatment with pemetrexed and MX, in the absence of immune cells. Concurrent knockdown of IDO and thymidylate synthase (TS, a key rate-limiting enzyme in DNA synthesis and repair) sensitizes human lung cancer cells to pemetrexed and 5FUdR to a greater degree than knockdown of either target alone. We conclude that BER in IDO-expressing A549 cells plays a major role in mediating resistance to a range of approved and candidate anticancer drugs. IDO inhibitors are undergoing clinical trials primarily to improve antitumor immune responses. We show that targeting IDO alone or in combination with TS is a potentially valuable therapeutic strategy for cancer treatment, independent of immune activity and in combination with conventional chemotherapy.


Subject(s)
Acrylamides/pharmacology , Deoxycytidine/analogs & derivatives , Down-Regulation/drug effects , Hydroxylamines/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Pemetrexed/pharmacology , Piperidines/pharmacology , Animals , BRCA2 Protein/metabolism , Cell Line, Tumor , Clone Cells , DNA Repair/drug effects , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Induction/drug effects , Floxuridine/pharmacology , Gene Knockdown Techniques , Humans , Mice, SCID , NAD/metabolism , RNA, Small Interfering/metabolism , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
6.
Oncotarget ; 6(26): 22397-409, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26087398

ABSTRACT

Nucleoside metabolism enzymes are determinants of chemotherapeutic drug activity. The nucleoside salvage enzyme deoxycytidine kinase (dCK) activates gemcitabine (2', 2'-difluoro-2'-deoxycytidine) and is negatively regulated by deoxycytidine triphosphate (dCTP). Reduction of dCTP in tumor cells could, therefore, enhance gemcitabine activity. Mitochondrial thymidine kinase 2 (TK2) phosphorylates deoxycytidine to generate dCTP. We hypothesized that: (1) TK2 modulates human tumor cell sensitivity to gemcitabine, and (2) antisense knockdown of TK2 would decrease dCTP and increase dCK activity and gemcitabine activation. siRNA downregulation of TK2 sensitized MCF7 and HeLa cells (high and moderate TK2) but not A549 cells (low TK2) to gemcitabine. Combined treatment with TK2 siRNA and gemcitabine increased dCK. We also hypothesized that TK2 siRNA-induced drug sensitization results in mitochondrial damage that enhances gemcitabine effectiveness. TK2 siRNA and gemcitabine decreased mitochondrial redox status, DNA content, and activity. This is the first demonstration of a direct role for TK2 in gemcitabine resistance, or any independent role in cancer drug resistance, and further distinguishes TK2 function from that of other dTMP-producing enzymes [cytosolic TK1 and thymidylate synthase (TS)]. siRNA knockdown of TK1 and/or TS did not sensitize cancer cells to gemcitabine indicating that, among the 3 enzymes, only TK2 is a candidate therapeutic target for combination with gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine Kinase/metabolism , Deoxycytidine/analogs & derivatives , Neoplasms/therapy , RNA, Small Interfering/administration & dosage , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Cell Line, Tumor , Deoxycytidine/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , MCF-7 Cells , Mitochondria/enzymology , Neoplasms/drug therapy , Neoplasms/genetics , RNA, Small Interfering/genetics , Transfection , Gemcitabine
7.
Mol Oncol ; 8(8): 1429-40, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24974076

ABSTRACT

Tumor cells have unstable genomes relative to non-tumor cells. Decreased DNA integrity resulting from tumor cell instability is important in generating favorable therapeutic indices, and intact DNA repair mediates resistance to therapy. Targeting DNA repair to promote the action of anti-cancer agents is therefore an attractive therapeutic strategy. BRCA2 is involved in homologous recombination repair. BRCA2 defects increase cancer risk but, paradoxically, cancer patients with BRCA2 mutations have better survival rates. We queried TCGA data and found that BRCA2 alterations led to increased survival in patients with ovarian and endometrial cancer. We developed a BRCA2-targeting second-generation antisense oligonucleotide (ASO), which sensitized human lung, ovarian, and breast cancer cells to cisplatin by as much as 60%. BRCA2 ASO treatment overcame acquired cisplatin resistance in head and neck cancer cells, but induced minimal cisplatin sensitivity in non-tumor cells. BRCA2 ASO plus cisplatin reduced respiration as an early event preceding cell death, concurrent with increased glucose uptake without a difference in glycolysis. BRCA2 ASO and cisplatin decreased metastatic frequency in vivo by 77%. These results implicate BRCA2 as a regulator of metastatic frequency and cellular metabolic response following cisplatin treatment. BRCA2 ASO, in combination with cisplatin, is a potential therapeutic anti-cancer agent.


Subject(s)
BRCA2 Protein/metabolism , Cisplatin/pharmacology , Animals , BRCA2 Protein/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Chick Embryo , Humans , Neoplasm Metastasis/genetics , Oligoribonucleotides, Antisense/pharmacology , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
8.
Oncotarget ; 5(9): 2778-91, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24784564

ABSTRACT

Indoleamine 2,3-dioxygenase-1 (IDO) is an immunosuppressive molecule expressed by most human tumors. IDO levels correlate with poor prognosis in cancer patients and IDO inhibitors are under investigation to enhance endogenous anticancer immunosurveillance. Little is known of immune-independent functions of IDO relevant to cancer therapy. We show, for the first time, that IDO mediates human tumor cell resistance to a PARP inhibitor (olaparib), gamma radiation, cisplatin, and combined treatment with olaparib and radiation, in the absence of immune cells. Antisense-mediated reduction of IDO, alone and (in a synthetic lethal approach) in combination with antisense to the DNA repair protein BRCA2 sensitizes human lung cancer cells to olaparib and cisplatin. Antisense reduction of IDO decreased NAD+ in human tumor cells. NAD+ is essential for PARP activity and these data suggest that IDO mediates treatment resistance independent of immunity and at least partially due to a previously unrecognized role for IDO in DNA repair. Furthermore, IDO levels correlated with accumulation of tumor cells in G1 and depletion of cells in G2/M of the cell cycle, suggesting that IDO effects on cell cycle may also modulate sensitivity to radiation and chemotherapeutic agents. IDO is a potentially valuable therapeutic target in cancer treatment, independent of immune function and in combination with other therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Gamma Rays , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Radiation Tolerance , BRCA2 Protein/antagonists & inhibitors , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Blotting, Western , Cell Cycle , Cell Proliferation , Cisplatin/administration & dosage , Combined Modality Therapy , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Neoplasms/pathology , Phthalazines/administration & dosage , Piperazines/administration & dosage , RNA, Small Interfering/genetics , Tumor Cells, Cultured
9.
Mol Ther Nucleic Acids ; 2: e78, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23481354

ABSTRACT

A high mutation rate leading to tumor cell heterogeneity is a driver of malignancy in human cancers. Paradoxically, however, genomic instability can also render tumors vulnerable to therapeutic attack. Thus, targeting DNA repair may induce an intolerable level of DNA damage in tumor cells. BRCA2 mediates homologous recombination repair, and BRCA2 polymorphisms increase cancer risk. However, tumors with BRCA2 mutations respond better to chemotherapy and are associated with improved patient prognosis. Thymidylate synthase (TS) is also involved in DNA maintenance and generates cellular thymidylate. We determined that antisense downregulation of BRCA2 synergistically potentiated drugs with mechanisms of action related to BRCA2 function (cisplatin, melphalan), a phenomenon we named "complementary lethality." TS knockdown induced complementary lethality to TS-targeting drugs (5-FUdR and pemetrexed) but not DNA cross-linking agents. Combined targeting of BRCA2 and TS induced complementary lethality to both DNA-damaging and TS-targeting agents, thus creating multidrug sensitive tumors. In addition, we demonstrated for the first time that simultaneous downregulation of both targets induced combined complementary lethality to multiple mechanistically different drugs in the same cell population. In this study, we propose and define the concept of "complementary lethality" and show that actively targeting BRCA2 and TS is of potential therapeutic benefit in multidrug treatment of human tumors. This work has contributed to the development of a BRCA2-targeting antisense oligdeoxynucleotide (ASO) "BR-1" which we will test in vivo in combination with our TS-targeting ASO "SARI 83" and attempt early clinical trials in the future.Molecular Therapy - Nucleic Acids (2013) 2, e78; doi:10.1038/mtna.2013.7 published online 12 March 2013.

10.
Cancer Lett ; 274(2): 279-89, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-18986763

ABSTRACT

N,N-diethyl-2-[4-(phenylmethyl)phenoxyl]ethanamine (tesmilifene), a tamoxifen derivative with antihistamine activity, greatly enhanced the survival of doxorubicin-treated, advanced stage breast cancer patients in a phase III trial. However, the molecular basis of tesmilifene action is not firmly established. The effects of tesmilifene on activity of several anticancer drugs was investigated using human head and neck squamous cell carcinoma (HNSCC) and breast carcinoma cell lines as a model system. Multidrug resistant (MDR) variants of an HNSCC cell line, HN-5a/V15e, and a breast carcinoma cell line, MCF-7/V25a, both highly overexpressed mdr1 (ABCB1) mRNA and the proteins P-glycoprotein and glutathione transferase-pi. Drug sensitivities were measured by a vital stain after 4 days of continuous exposure to anticancer drug in the absence and presence of tesmilifene at a concentration that alone had no antiproliferative effect. Tesmilifene had minimal effect on drug cytotoxicity against the parental cell lines. However, the same tesmilifene treatment enhanced cytotoxicity of docetaxel, paclitaxel, epirubicin, doxorubicin, and vinorelbine against both MDR cell lines by up to 50%. Flow cytometric measurement of annexin V/propidium iodide staining demonstrated that tesmilifene increased the killing of HN-5a/V15e cells caused by docetaxel after 24 and 48h exposure. Tesmilifene increased accumulation of radiolabelled vincristine in HN-5a/V15e cells, over 4h, by up to 100%. The results suggest that tesmilifene might be effective in the treatment of tumors that are resistant to natural product drugs. The mechanism of enhancement appears to be related to expression of an ABC pump-dependent, MDR phenotype.


Subject(s)
Biological Products/pharmacology , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Histamine Antagonists/pharmacology , Phenyl Ethers/pharmacology , Apoptosis , Base Sequence , Cell Line, Tumor , DNA Primers , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Flow Cytometry , Genes, MDR , Humans
11.
DNA Cell Biol ; 27(5): 229-40, 2008 May.
Article in English | MEDLINE | ID: mdl-18358073

ABSTRACT

Thymidylate synthase (TS) is essential for DNA replication and is a target for cancer chemotherapy. However, toxicity to normal cells and tumor cell drug resistance necessitate development of new therapeutic strategies. One such strategy is to use antisense (AS) technology to reduce TS mRNA and protein levels in treated cells. We have developed oligodeoxynucleotides (ODNs) that target different regions of TS mRNA, inhibit human tumor cell proliferation as single agents, and enhance cytotoxicity of clinically useful TS protein-targeting drugs. Here we describe ODN 491, a novel 20mer AS ODN complementary to a previously untargeted portion of the TS mRNA coding region. AS ODN 491 decreased TS mRNA levels to different degrees in a panel of human tumor-derived cell lines, and induced different physiological effects in a tumor cell line-dependent manner. ODN 491 (like AS TS ODN 83, previously shown to be effective) decreased TS protein levels in HeLa cells with a concomitant increase in sensitivity to TS-targeting chemotherapeutics. However (and contrary to HeLa cell response to an AS ODN 83), it did not, as a single agent, inhibit HeLa cell proliferation. In MCF-7 cells, ODN 491 treatment was less effective at reducing TS mRNA and did not reduce TS protein, nor did it enhance sensitivity to TS-targeting or other chemotherapeutics. Moreover, specifically in MCF-7 cells but not HeLa cells, ODN 491 as a single agent induced apoptosis. These data indicate that AS TS ODN 491 is an effective AS reagent targeting a novel TS mRNA region. However, treatment of tumor cell lines with AS TS ODNs targeting different TS mRNA regions results in a pattern of physiological effects that varies in a tumor cell line-specific fashion. In addition, the capacity of different AS TS ODNs to induce physiological effects does not correlate well with their capacity to reduce TS mRNA and/or protein and, further, depends on the region of TS mRNA selected for targeting. Recognition of tumor cell-specific and mRNA region-specific variability in response to AS TS ODNs will be important in designing AS TS ODNs for potential clinical use.


Subject(s)
Antineoplastic Agents/pharmacology , Oligonucleotides, Antisense/pharmacology , Thymidylate Synthase/genetics , Apoptosis/physiology , Cell Proliferation/drug effects , Flow Cytometry , HeLa Cells/drug effects , Humans , RNA, Messenger/metabolism , Transcription, Genetic , Transfection
12.
Cancer Lett ; 251(1): 168-78, 2007 Jun 18.
Article in English | MEDLINE | ID: mdl-17197076

ABSTRACT

Tangeretin and nobiletin are citrus flavonoids that are among the most effective at inhibiting cancer cell growth in vitro and in vivo. The antiproliferative activity of tangeretin and nobiletin was investigated in human breast cancer cell lines MDA-MB-435 and MCF-7 and human colon cancer line HT-29. Both flavonoids inhibited proliferation in a dose- and time-dependent manner, and blocked cell cycle progression at G1 in all three cell lines. At concentrations that resulted in significant inhibition of proliferation and cell cycle arrest, neither flavonoid induced apoptosis or cell death in any of the tumor cell lines. To test the ability of arrested cells to recover, cells that were incubated with tangeretin and nobiletin for 4 days were then cultured in flavonoid-free medium for an additional 4 days. Cells resumed proliferation similar to untreated control within a day of flavonoid removal. Cell cycle distribution was similar to that of control within 4 days of flavonoid removal. These data indicate that, in these cell lines at concentrations that inhibit proliferation up to 80% over 4 days, tangeretin and nobiletin are cytostatic and significantly suppress proliferation by cell cycle arrest without apoptosis. Such an agent could be expected to spare normal tissues from toxic side effects. Thus, tangeretin and nobiletin could be effective cytostatic anticancer agents. Inhibition of proliferation of human cancers without inducing cell death may be advantageous in treating tumors as it would restrict proliferation in a manner less likely to induce cytotoxicity and death in normal, non-tumor tissues.


Subject(s)
Apoptosis/drug effects , Flavones/pharmacology , G1 Phase/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Colonic Neoplasms/physiopathology , Dose-Response Relationship, Drug , Female , Flavones/chemistry , HT29 Cells , Humans , Molecular Structure , Time Factors
13.
Nutr Cancer ; 56(1): 86-94, 2006.
Article in English | MEDLINE | ID: mdl-17176222

ABSTRACT

Edible fruits and berries may serve as sources for novel anticancer agents, given that extracts of these foods have demonstrated cytotoxic activity against tumor cell lines. Semipurified, flavonoid-rich extracts of cranberry (Vaccinia macrocarpa) were shown previously to arrest proliferation of tumor cells and induce apoptosis. However, the ability of cranberry flavonoids to inhibit tumor growth in vivo has not been reported other than in a preliminary report. As model systems for testing this activity, human tumor cell lines representative of three malignancies were chosen: glioblastoma multiforme (U87), colon carcinoma (HT-29), and androgen-independent prostate carcinoma (DU145). A flavonoid-rich fraction 6 (Fr6) and a more purified proanthocyanidin (PAC)-rich fraction were isolated from cranberry presscake and whole cranberry, respectively, by column chromatography. Fr6 and PAC each significantly slowed the growth of explant tumors of U87 in vivo, and PAC inhibited growth of HT-29 and DU145 explants (P < 0.05), inducing complete regression of two DU145 tumor explants. Flow cytometric analyses of in vitro-treated U87 cells indicated that Fr6 and PAC could arrest cells in G1 phase of the cell cycle (P < 0.05) and also induce cell death within 24 to 48 h of exposure (P < 0.05). These results indicate the presence of a potential anticancer constituent in the flavonoid-containing fractions from cranberry extracts.


Subject(s)
Cell Line, Tumor/drug effects , Flavonoids/pharmacology , Plant Extracts/pharmacology , Vaccinium macrocarpon/chemistry , Animals , Apoptosis/drug effects , Biological Assay , Cell Division/drug effects , Colonic Neoplasms/pathology , Female , Flow Cytometry , G1 Phase/drug effects , Glioblastoma/pathology , HT29 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Prostatic Neoplasms/pathology , Time Factors
14.
J Nutr ; 134(6): 1529-35, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15173424

ABSTRACT

In light of the continuing need for effective anticancer agents, and the association of fruit and vegetable consumption with reduced cancer risk, edible plants are increasingly being considered as sources of anticancer drugs. Cranberry presscake (the material remaining after squeezing juice from the berries), when fed to mice bearing human breast tumor MDA-MB-435 cells, was shown previously to decrease the growth and metastasis of tumors. Therefore, further studies were undertaken to isolate the components of cranberry that contributed to this anticancer activity, and determine the mechanisms by which they inhibited proliferation. Using standard chromatographic techniques, a warm-water extract of cranberry presscake was fractionated, and an acidified methanol eluate (Fraction 6, or Fr6) containing flavonoids demonstrated antiproliferative activity. The extract inhibited proliferation of 8 human tumor cell lines of multiple origins. The androgen-dependent prostate cell line LNCaP was the most sensitive of those tested (10 mg/L Fr6 inhibited its growth by 50%), and the estrogen-independent breast line MDA-MB-435 and the androgen-independent prostate line DU145 were the least sensitive (250 mg/L Fr6 inhibited their growth by 50%). Other human tumor lines originating from breast (MCF-7), skin (SK-MEL-5), colon (HT-29), lung (DMS114), and brain (U87) had intermediate sensitivity to Fr6. Using flow cytometric analyses of DNA distribution (cell cycle) and annexin V-positivity (apoptosis), Fr6 was shown in MDA-MB-435 cells to block cell cycle progression (P < 0.05) and induce cells to undergo apoptosis (P < 0.05) in a dose-dependent manner. Fr6 is potentially a source of a novel anticancer agent.


Subject(s)
Cell Line, Tumor/pathology , Flavonoids/pharmacology , Plant Extracts/pharmacology , Vaccinium macrocarpon/chemistry , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Chemical Fractionation , Female , Humans , Male , Prostatic Neoplasms/pathology
15.
Cancer Gene Ther ; 10(4): 278-86, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12679800

ABSTRACT

Thymidylate synthase (TS) catalyzes de novo production of thymidylate for DNA synthesis and cell proliferation. As such, TS has been a target of antitumor chemotherapy for many years. Our laboratory has identified several antisense oligodeoxynucleotides (ODNs) that downregulate TS mRNA and protein, inhibit cell proliferation, and sensitize cells to TS-directed chemotherapeutic drugs. Based on our observation that targeting distinct regions of the TS mRNA with a variety of antisense molecules resulted in differential effects on TS mRNA levels, it was hypothesized that use of multiple ODNs targeting distinct noncontiguous regions would result in synergistic or antagonistic interactions. In this study, we report that some combinations of TS antisense ODNs were more effective at reducing TS mRNA abundance and inhibiting cell proliferation than the individual ODNs used alone. However, in contrast to the effects on cell proliferation, the enhanced sensitivity to anti-TS chemotherapeutic drugs (i.e., raltitrexed and 5-fluorodeoxyuridine) that is achieved by treatment with individual ODNs was not further augmented by combined ODN treatment. This suggests that ODNs targeting TS mRNA inhibit an alternative function of TS mRNA or protein, distinct from thymidylate production. The results are evidence that the novel use of multiple antisense ODNs that target different regions of the same mRNA represents a general strategy to improve antisense effectiveness.


Subject(s)
Antineoplastic Agents , Neoplasms/therapy , Oligodeoxyribonucleotides, Antisense/genetics , Thymidylate Synthase/genetics , Antineoplastic Agents/therapeutic use , Cell Division , Down-Regulation , Drug Resistance, Neoplasm , Female , Fluorouracil/therapeutic use , HeLa Cells , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Quinazolines/therapeutic use , RNA, Messenger/analysis , Thiophenes/therapeutic use , Thymidylate Synthase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...