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










Publication year range
1.
Wiad Lek ; 76(12): 2543-2555, 2023.
Article in English | MEDLINE | ID: mdl-38290016

ABSTRACT

Marie Sklodowska-Curie Symposia on Cancer Research and Care (MSCS-CRC) promote collaborations between cancer researchers and care providers in the United States, Canada and Central and Eastern European Countries (CEEC), to accelerate the development of new cancer therapies, advance early detection and prevention, increase cancer awareness, and improve cancer care and the quality of life of patients and their families. The third edition of MSCS-CRC, held at Roswell Park Comprehensive Cancer Center, Buffalo, NY, in September 2023, brought together 137 participants from 20 academic institutions in the US, Poland, Ukraine, Lithuania, Croatia and Hungary, together with 16 biotech and pharma entities. The key areas of collaborative opportunity identified during the meeting are a) creating of a database of available collaborative projects in the areas of early-phase clinical trials, preclinical development, and identification of early biomarkers; b) promoting awareness of cancer risks and efforts at cancer prevention; c) laboratory and clinical training; and d) sharing experience in cost-effective delivery of cancer care and improving the quality of life of cancer patients and their families. Examples of ongoing international collaborations in the above areas were discussed. Participation of the representatives of the Warsaw-based Medical Research Agency, National Cancer Institute (NCI) of the United States, National Cancer Research Institutes of Poland and Lithuania, New York State Empire State Development, Ministry of Health of Ukraine and Translational Research Cancer Center Consortium of 13 cancer centers from the US and Canada, facilitated the discussion of available governmental and non-governmental funding initiatives in the above areas.


Subject(s)
Biomedical Research , Neoplasms , Humans , United States , New York , Quality of Life , Neoplasms/therapy , Poland
2.
Cancer Lett ; 311(2): 219-29, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-21872389

ABSTRACT

The goal of this study is to determine whether treatment with methylselenocysteine (MSC) results in differential uptake of irinotecan and its active metabolite (SN-38) between tumors of head and neck squamous cell carcinomas and normal tissue. The in vivo synergy between MSC and irinotecan is influenced by treatment schedule and associated with enhancement of tumor vessel maturation, intra-tumor concentration of SN-38 and apoptotic death of tumor cells. Normal tissue drug concentrations were not impacted by selenium treatment. The finding is of clinical relevance for enabling the delivery of higher doses of irinotecan to reverse tumor resistance, recurrence and ultimately enhancing cure rates.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Camptothecin/analogs & derivatives , Animals , Apoptosis/drug effects , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Chromatography, High Pressure Liquid , Cysteine/administration & dosage , Cysteine/analogs & derivatives , Cysteine/pharmacokinetics , Drug Synergism , Female , Humans , Immunohistochemistry , Irinotecan , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Organoselenium Compounds/administration & dosage , Organoselenium Compounds/pharmacokinetics , Selenocysteine/analogs & derivatives , Xenograft Model Antitumor Assays
3.
Prostate ; 71(11): 1178-88, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21656828

ABSTRACT

BACKGROUND: Reduced expression of prostate-derived Ets transcription factor (PDEF) leads to morphologic change as well as increased migration and invasiveness of prostate cancer cells. However, the clinical relevance of PDEF expression and its relationship to anti-apoptotic protein survivin is yet to be determined. METHODS: Tissue microarrays of 73 prostate carcinomas and their adjacent benign prostate tissue, as well as 50 benign prostates were evaluated for PDEF expression by immunohistochemistry. Results were confirmed in available tumor tissues using Western blot and RT-PCR. Expression of survivin in prostate carcinoma and benign tissues were determined using Western blot. Results and correlation with clinical data were statistically analyzed. RESULTS: Patients' specimens with low Gleason scores (GS < 5) expressed higher levels of PDEF protein and lower levels of survivin protein when compared with moderate-to-high GS tumors (GS > 6). Patients with PDEF-positive tumor survived significantly longer (P < 0.0001) than patients with PDEF-negative tumor, and the 8-year survival rate was 94% and 40%, respectively. PDEF expression was detected at the highest levels in benign tissues and was down-regulated or lost in 30 recently diagnosed prostate carcinomas. Re-expression of PDEF in prostate cancer cells inhibited survivin expression. Treatment of prostate cancer cells with methylseleninic acid resulted in restoration of PDEF expression, down-regulation of survivin, and inhibition of tumor cell growth when compared with untreated controls (P < 0.05). CONCLUSIONS: These studies demonstrated an inverse correlation between PDEF and survivin expression, and that up-regulation of PDEF was associated with a favorable prognosis in patients with clinically localized prostate cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-ets/biosynthesis , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Male , Prognosis , RNA, Messenger/biosynthesis , Survivin
4.
Mol Cancer ; 9: 310, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21126356

ABSTRACT

BACKGROUND: Beta-catenin is a multifunctional oncogenic protein that contributes fundamentally to cell development and biology. Elevation in expression and activity of ß-catenin has been implicated in many cancers and associated with poor prognosis. Beta-catenin is degraded in the cytoplasm by glycogen synthase kinase 3 beta (GSK-3ß) through phosphorylation. Cell growth and proliferation is associated with ß-catenin translocation from the cytoplasm into the nucleus. This laboratory was the first to demonstrate that selenium-containing compounds can enhance the efficacy and cytotoxicity of anticancer drugs in several preclinical xenograft models. These data provided the basis to identify mechanism of selenium action focusing on ß-catenin as a target. This study was designed to: (1) determine whether pharmacological doses of methylseleninic acid (MSeA) have inhibitory effects on the level and the oncogenic activity of ß-catenin, (2) investigate the kinetics and the mechanism of ß-catenin inhibition, and (3) confirm that inhibition of ß-catenin would lead to enhanced cytotoxicity of standard chemotherapeutic drugs. RESULTS: In six human cancer cell lines, the inhibition of total and nuclear expression of ß-catenin by MSeA was dose and time dependent. The involvement of GSK-3ß in the degradation of ß-catenin was cell type dependent (GSK-3ß-dependent in HT-29, whereas GSK-3ß-independent in HCT-8). However, the pronounced inhibition of ß-catenin by MSeA was independent of various drug treatments and was not reversed after combination therapy.Knockout of ß-catenin by ShRNA and its inhibition by MSeA yielded similar enhancement of cytotoxicity of anticancer drugs.Collectively, the generated data demonstrate that ß-catenin is a target of MSeA and its inhibition resulted in enhanced cytotoxicity of chemotherapeutic drugs. CONCLUSIONS: This study demonstrates that ß-catenin, a molecule associated with drug resistance, is a target of selenium and its inhibition is associated with increased multiple drugs cytotoxicity in various human cancers. Further, degradation of ß-catenin by GSK-3ß is not a general mechanism but is cell type dependent.


Subject(s)
Neoplasms/metabolism , beta Catenin/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Humans , Organoplatinum Compounds/pharmacology , Organoselenium Compounds/pharmacology , Oxaliplatin , Paclitaxel/pharmacology , RNA Interference , Taxoids/pharmacology , Topotecan/pharmacology , beta Catenin/genetics
5.
Am J Transl Res ; 1(4): 393-405, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19956451

ABSTRACT

Growing evidence indicates that the antiapoptotic protein survivin is a major factor of drug and radiation resistance in cancer cells. However, application of this finding to therapeutic drug combination is largely unexplored. In this study, breast cancer cells were used for treatment with anticancer compounds alone or in combination. We report that T138067, a better drug against multiple drug resistance (MDR) tumor cells than taxol (Shan et al., PNAS 96:5686-91,1999), induces survivin expression and consequently decreases its effectiveness on the induction of cancer cell death. Treatment of breast cancer cells with T138067 induced survivin expression in these cells while showing no effect on Bcl-2, indicating its specificity. Upregulation of survivin by T138067 was concomitant with an increased drug resistance and associated with an increased phosphorylation of Akt and Erk1/2 MAPK, and a decreased phosphorylation of p38 MAPK without affecting the phosphorylation of ErbB2. Therefore, it is possible that inhibition of T138067-induced survivin expression by alternative approaches may sensitize cells to T138067-induced cell death. We found that treatment of breast cancer cells with SN38, the active metabolite of irinotecan, inhibits survivin expression. Intriguingly, inhibition of survivin expression by SN38 was more effective at a low concentration than at the high concentration, which makes SN38 a good survivin modulator. Furthermore, in contrast with the decreased phosphorylation of p38 MAPK after T138067 treatment, inhibition of survivin expression by SN38 was associated with an increased phosphorylation of the p38 MAPK, suggesting opposing signals converging to survivin. Consistent with these observations, T138067 in combination with SN38 strongly induced cell death in comparison with each drug alone. Similarly, sequential combination of resveratrol, a component of red grapes that inhibits survivin expression, with T138067 also provoked massive breast cancer cell death compared with T138067 alone. Together, these results highlight a new concept that unique signaling cross talk converged to survivin may be considered for rational drug combination in the clinic.

6.
Cancer Biol Ther ; 7(12): 1901-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18981709

ABSTRACT

This study evaluates methylseleninic acid (MSeA) improvement of paclitaxel efficacy against human ovarian cancer (skov3) with regard to survivin expression. MSeA and paclitaxel alone and in concurrent or sequential combination treatments were tested. Cell growth/death was evaluated using SRB, trypan blue, colony formation and ELISA assays. Cells were transfected with survivin shRNA and survivin's expression was measured using RT-PCR and Western blots. Drugs interaction was further evaluated using isobologram analyses. Different treatments with MSeA did not enhance paclitaxel's efficacy in the wild type skov3. Silencing survivin had no effect on MSeA or paclitaxel efficacy when used alone or in concurrent combination. After sequential combination treatment, synergy and significant induction of apoptosis were observed in cells transfected with survivin shRNA. However, antagonism and minimal induction of apoptosis were observed in empty or scramble survivin shRNA transfected cells. In conclusion, these data suggest that synergy between MSeA and paclitaxel in skov3 is associated with silencing survivin expression.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Microtubule-Associated Proteins/genetics , Organoselenium Compounds/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Paclitaxel/therapeutic use , Cell Death/drug effects , Cell Division/drug effects , Drug Synergism , Female , Humans , Inhibitor of Apoptosis Proteins , Ovarian Neoplasms/pathology , Survivin
7.
Int J Cancer ; 123(6): 1376-84, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18567002

ABSTRACT

We have previously shown that ovarian tumors express prostate-derived Ets transcription factor (PDEF). However, the precise role of PDEF in the prognosis of ovarian cancer is unknown. In our study, we report for the first time that expression of PDEF in tumor lesions of patients with ovarian cancer is associated with favorable prognosis. Evaluation of samples from 40 patients with ovarian cancer showed that early stage (IA) and borderline (IIB, III) ovarian tumors expressed higher levels of PDEF mRNA and protein and lower levels of survivin compared to late stage ovarian tumors (IIIC and IV, p < 0.05). Normal ovarian tissues expressed the highest levels of PDEF mRNA and protein when compared to tumor tissues (p < 0.05). A Log-Rank test showed that overall survival of patients with PDEF-positive and survivin-negative ovarian tumors was significantly longer than those with PDEF-negative and survivin-positive tumors (p < 0.01). Forced expression of PDEF in PDEF-negative ovarian tumor cells inhibited tumor cell growth, induced apoptosis, downregulated survivin expression and its promoter activity. Furthermore, treatment of ovarian cancer cells with vitamin D or a selenium compound resulted in re-expression of PDEF, downregulation of survivin, induction of apoptosis and inhibition of tumor cell growth when compared to untreated controls (p < 0.05). Together, these observations showed an inverse correlation between PDEF and survivin expression and suggested that increased PDEF expression along with reduced survivin was associated with prolonged survival of patients with ovarian cancer.


Subject(s)
Biomarkers, Tumor/analysis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-ets/biosynthesis , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Kaplan-Meier Estimate , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasm Staging , Ovarian Neoplasms/mortality , Prognosis , Proto-Oncogene Proteins c-ets/drug effects , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Survivin
8.
Biochem Pharmacol ; 73(9): 1280-7, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17239826

ABSTRACT

This study was designed to understand the basis for the efficacy of methylselenocysteine (MSC) in increasing the therapeutic index of irinotecan against human tumor xenografts. Nude mice bearing human head and neck squamous cells carcinoma xenografts (FaDu and A253) were treated orally with different doses of MSC and irinotecan. Plasma, tumor and normal tissue samples were collected at different times after MSC treatments and were analyzed for selenium (Se) concentration using electrothermal atomic absorption spectrophotometry. MSC is highly effective in modulating the therapeutic index of irinotecan. Enhanced irinotecan efficacy was greater in FaDu tumors (100% CR) than in A253 tumors (60% CR), and depended on MSC dose with a minimum effective dose of 0.01 mg/dx28. The highest plasma Se concentration was achieved 1h after a single dose and 28 d after daily treatments of MSC. The ability of FaDu tumors to retain Se was significantly better than A253 tumors, and the highest Se concentration in normal tissue was achieved in the liver. Peak plasma and tissue Se concentrations were functions of the dose and duration of MSC treatment. The MSC-dependent increase in Se level in normal tissues may contribute to the protective effect against irinotecan toxicity observed in those tissues. Intratumoral total Se concentration was not found to be predictive of the combination therapy response rates. There is a critical need to develop a method to measure the active metabolite of MSC, rather than total Se.


Subject(s)
Camptothecin/analogs & derivatives , Cysteine/analogs & derivatives , Neoplasms/drug therapy , Organoselenium Compounds/pharmacology , Plasma/drug effects , Selenium/blood , Administration, Oral , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cysteine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Irinotecan , Kidney/drug effects , Kidney/metabolism , Kinetics , Liver/drug effects , Liver/metabolism , Mice , Mice, Nude , Plasma/chemistry , Plasma/metabolism , Selenium/metabolism , Selenium/pharmacokinetics , Selenocysteine/analogs & derivatives
9.
Mol Cancer Ther ; 5(10): 2540-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17041098

ABSTRACT

The study was designed to evaluate the combination treatment of methylselenocysteine (MSeC) and docetaxel and to delineate the underlying mechanism associated with observed in vitro synergy between MSeC and docetaxel in prostate cancer cells. Cells were treated with different concentrations and schedules (concurrent or sequential) of MSeC and docetaxel alone or in combination. Cell growth/death was assessed with sulforhodamine B assay, trypan blue assay, and time-lapse video. Loewe synergism/antagonism model was used to determine whether the combination effect was additive, synergistic, or antagonistic. Apoptosis and caspase-3 activity were evaluated with cell death ELISA assay and caspase activity assay, respectively. Synergy between MSeC and docetaxel was further assessed in the presence and absence of z-VAD-fmk, a pan-caspase inhibitor. Effect of MSeC and docetaxel alone or in combination on the cellular expression of the antiapoptotic protein survivin was measured with Western blot analyses. Pretreatment with MSeC was crucial to enhance docetaxel antitumor activity. The enhanced antitumor activity of the sequential combination treatment of MSeC and docetaxel (MSeC/docetaxel) was highly synergistic. Apoptosis increased after MSeC/docetaxel, compared with each drug alone or concurrent treatment. Pretreatment with z-VAD-fmk converted the synergy into antagonism, suggesting that the synergy is caspase-dependent apoptosis. The survivin level was down-regulated following MSeC/docetaxel treatment when compared with each drug alone. In conclusion, pretreatment with MSeC was essential to markedly sensitize cells to docetaxel. The synergy between MSeC and docetaxel in C2G prostate cancer cells is associated with increased level of caspase-dependent apoptosis and decreased level of survivin.


Subject(s)
Antineoplastic Agents/pharmacology , Cysteine/analogs & derivatives , Organoselenium Compounds/pharmacology , Taxoids/pharmacology , Animals , Anticarcinogenic Agents/pharmacology , Apoptosis , Caspase 3/metabolism , Cell Line, Tumor , Cysteine/pharmacology , Docetaxel , Down-Regulation , Drug Synergism , Enzyme Activation , Inhibitor of Apoptosis Proteins , Male , Mice , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms , Selenocysteine/analogs & derivatives , Survivin
10.
Clin Cancer Res ; 12(4): 1237-44, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489079

ABSTRACT

PURPOSE: We conducted a phase I study to determine the maximum tolerated dose (MTD) of irinotecan with fixed, nontoxic high dose of selenomethionine. EXPERIMENTAL DESIGN: Selenomethionine was given orally as a single daily dose containing 2,200 mug of elemental selenium (Se) starting 1 week before the first dose of irinotecan. Irinotecan was given i.v. once weekly x 4 every 6 weeks (one cycle). The starting dose of irinotecan was 125 mg/m(2)/wk. Escalation occurred in cohorts of three patients until the MTD was defined. Pharmacokinetic studies were done for selenium and irinotecan and its metabolites. RESULTS: Three of four evaluable patients at dose level 2 of irinotecan (160 mg/m(2)/wk) had a dose-limiting diarrhea. None of the six evaluable patients at dose level 1 (125 mg/m(2)/wk irinotecan) had a dose-limiting toxicity. One patient with history of irinotecan-refractory colon cancer achieved a partial response. The long half-life of selenium resulted in a prolonged accumulation towards steady-state concentrations. No significant changes in the pharmacokinetics of CPT-11, SN-38, or SN-38G were identified; however, the coadministration of selenomethionine significantly reduced the irinotecan biliary index, which has been associated with gastrointestinal toxicity. CONCLUSIONS: Selenomethionine at 2,200 mug/d did not allow the safe escalation of irinotecan beyond the previously defined MTD of 125 mg/m(2). None of the patients receiving 125 mg/m(2) of irinotecan had grade >2 diarrhea. Unexpected responses and disease stabilizations were noted in a highly refractory population. Further escalation of selenomethionine is recommended in future trials to achieve defined protective serum concentrations of selenium.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Area Under Curve , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Female , Humans , Injections, Intravenous , Irinotecan , Male , Middle Aged , Nausea/chemically induced , Neoplasms/metabolism , Selenomethionine/administration & dosage , Selenomethionine/adverse effects , Selenomethionine/pharmacokinetics , Treatment Outcome
11.
Mol Cancer Ther ; 4(5): 843-54, 2005 May.
Article in English | MEDLINE | ID: mdl-15897249

ABSTRACT

The combination of methylselenocysteine and irinotecan (CPT-11) is synergistic against FaDu and A253 xenografts. Methylselenocysteine/CPT-11 increased tumor cure rate to 100% in FaDu and to 60% in A253. In this study, the effect of methylselenocysteine on pharmacokinetic and pharmacogenetic profiles of genes relevant to CPT-11 metabolic pathway was evaluated to identify possible mechanisms associated with the observed combinational synergy. Nude mice bearing tumors (FaDu and A253) were treated with methylselenocysteine, CPT-11, and a combination of methylselenocysteine/CPT-11. Samples were collected and analyzed for plasma and intratumor concentration of CPT-11 and 7-ethyl-10-hydroxyl-camptothecin (SN-38) by high-performance liquid chromatography. The intratumor relative expression of genes related to the CPT-11 metabolic pathway was measured by real-time PCR. After methylselenocysteine treatment, the intratumor area under the concentration-time curve of SN-38 increased to a significantly higher level in A253 than in FaDu and was associated with increased expression of CES1 in both tumors. Methylselenocysteine/CPT-11 treatment, compared with CPT-11 alone, resulted in a significant decrease in levels of ABCC1 and DRG1 in FaDu tumors and an increase in levels of CYP3A5 and TNFSF6 in A253 tumors. No statistically significant changes induced by methylselenocysteine/CPT-11 were observed in the levels of other investigated variables. In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. No correlation was observed between cure rate and other investigated variables (transporters, degradation enzymes, DNA repair, and cell survival/death genes) in either tumor.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Biomarkers, Tumor/metabolism , Camptothecin/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Cysteine/analogs & derivatives , Head and Neck Neoplasms/drug therapy , Pharmacogenetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Camptothecin/administration & dosage , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/genetics , Cysteine/administration & dosage , Female , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/genetics , Humans , Irinotecan , Mice , Mice, Nude , Organoselenium Compounds/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Selenocysteine/analogs & derivatives , Transplantation, Heterologous , Tumor Cells, Cultured
12.
Clin Cancer Res ; 10(23): 8005-17, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15585636

ABSTRACT

PURPOSE: Combination chemotherapy with irinotecan (CPT-11; 50 mg/kg/week x 4 intravenously), followed 24 hour later by 5-fluorouracil (50 mg/kg/week x 4 intravenously), results in 10 and 100% cure rates of animals bearing human head and neck squamous cell carcinoma xenografts A253 and FaDu, respectively. A253 consists of 30% well-differentiated and avascular and 70% poorly differentiated regions with low microvessel density (10/x400), whereas FaDu is uniformly poorly differentiated with higher microvessel density (19/x400). Studies were carried out for determining the role of well-differentiated and avascular regions in drug resistance in A253 and detection of such regions with noninvasive functional magnetic resonance (fMR) imaging. EXPERIMENTAL DESIGN: Tumors were harvested for histopathologic evaluation and immunohistochemistry (CD31, CD34; differentiation marker: involucrin; hypoxia markers: carbonic anhydrase IX, pimonidazole; vascular endothelial factor (VEGF) and Ki67) immediately after fMR imaging following the 3rd dose of chemotherapy. High-performance liquid chromatography determination of intratumoral drug concentration of 7-ethyl-10-hydroxyl-camptothecin and autoradiography with (14)C-labeled CPT-11 was done 2 hours after CPT-11 administration. RESULTS: Although A253 xenografts showed three times higher concentration of 7-ethyl-10-hydroxyl-camptothecin, FaDu was more responsive to therapy. After therapy, A253 tumor consisted mostly (approximately 80%) of well-differentiated regions (positive for involucrin) lacking microvessels with a hypoxic rim (positive for carbonic anhydrase IX and pimonidazole) containing few proliferating (Ki67 positive) poorly differentiated cells. Autoradiography revealed that well-differentiated A253 tumor regions showed 5-fold lower (14)C-labeled CPT-11 concentrations compared with poorly differentiated areas (P < 0.001). Blood oxygen level dependant fMR imaging was able to noninvasively distinguish the hypoxic and well-vascularized regions within the tumors. CONCLUSION: Avascular-differentiated regions in squamous cell carcinoma offer sanctuary to some hypoxic but viable tumor cells (carbonic anhydrase IX and Ki67 positive) that escape therapy because of limited drug delivery. This study provides direct evidence that because of a specific histologic structure, avascular, well-differentiated hypoxic regions in tumors exhibit low drug uptake and represent a unique form of drug resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Cell Differentiation , Drug Resistance, Neoplasm , Head and Neck Neoplasms/drug therapy , Hypoxia , Animals , Biomarkers, Tumor/metabolism , Camptothecin/administration & dosage , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/radiotherapy , Drug Delivery Systems , Female , Fluorouracil/administration & dosage , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/radiotherapy , Humans , Irinotecan , Magnetic Resonance Imaging , Mice , Mice, Nude , Microcirculation , Oxygen/blood , Rats , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Mol Pharmacol ; 66(1): 153-60, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15213307

ABSTRACT

Methylselenocysteine (MSC) is an organic selenium compound in preventative clinical trials involving prostate, lung, and colon carcinoma. We found that methioninase-activated MSC potentiates 7-ethyl-10-hydroxycamptothecin (SN-38)-induced cell lethality in vitro in the p53-defective human head and neck carcinoma A253 cells. Activated MSC increases chk2 phosphorylation at threonine-68 induced by SN-38, with no significant effect on chk1 phosphorylation. Cell cycle arrest induced by SN-38, however, was not abrogated or potentiated by MSC. These results suggest that the enhanced cellular lethality of SN-38 by MSC was not associated with cell cycle regulation pathways. Because chk2, in addition to its role in cell cycle arrest, can induce apoptosis by phosphorylation/activation, we examined whether increased chk2 phosphorylation could induce preapoptotic DNA fragmentation. DNA damage analysis showed that megabase DNA fragmentation is decreased, accompanied by the increased 30 to 300 kilobase pairs of DNA fragmentation after exposure to SN-38 with MSC, compared with SN-38 alone. No significant changes in the amount of DNA fragments were observed in cells treated with SN-38 or MSC alone. Moreover, proteolytic destruction of DNA replication-associated proteins cdc6, MCM2, and cdc25A may induce a DNA damage checkpoint response. The observed down-regulation of DNA replication proteins cdc6, MCM2, and cdc25A after exposure to SN-38 with MSC further indicates a relationship between drug response and DNA damage. Exposure to SN-38 with MSC resulted in a significant increase of poly(ADP-ribose) polymerasecleavage and caspase 3 activation. All together, the data support the hypothesis that enhanced lethality of this combination is associated with increased chk2 phosphorylation at Thr68 and down-regulation of specific DNA replication-associated proteins, which result in poly(ADP-ribose) polymerase cleavage, caspase 3 activation, and the induction of 30 to 300 kilobase pairs of DNA fragmentation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cysteine/analogs & derivatives , Cysteine/pharmacology , Organoselenium Compounds/pharmacology , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Carbon-Sulfur Lyases/metabolism , Caspase 3 , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Fragmentation/drug effects , Drug Interactions , Enzyme Activation , Gene Expression/drug effects , Humans , Irinotecan , Minichromosome Maintenance Complex Component 2 , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinases/metabolism , Selenocysteine/analogs & derivatives , cdc25 Phosphatases/metabolism
14.
Clin Cancer Res ; 10(3): 1121-9, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871992

ABSTRACT

PURPOSE: Although the combination of irinotecan and 5-Fluorouracil is clinically active, it is associated with significant toxicity and resistance. Studies were carried out to define the optimal dosage, sequence, and timing for the combination in mice bearing xenografted human tumors. EXPERIMENTAL DESIGN: The maximum tolerated dose of irinotecan and 5-Fluorouracil in combination was determined in nude mice. Therapeutic efficacy against established human colon carcinoma xenografts, HCT-8 and HT-29, and human head and neck squamous cell carcinoma xenografts, FaDu and A253, was determined using the rugs individually, simultaneously, and in sequence with various intervals in between. Treatments were i.v. weekly x 4. Immunohistochemical and reverse transcription-PCR measurements of relevant drug-metabolizing enzymes, apoptosis-related proteins, cell cycle distribution, cyclin A, and S phase fraction expression were carried out and compared with the therapeutic outcome. RESULTS: The maximum tolerated dose of irinotecan resulted in cure rates of 30% or less in all xenografts. No cures were achieved with FUra alone. Concurrent administration of irinotecan and FUra, or of FUra 24 h before irinotecan, resulted in cure rates of <20%, except for FaDu (60%). Administration of irinotecan 24 h before FUra resulted in the highest cure rates, 80% in HCT-8, 0% in HT-29, 100% in FaDu, and 10% in A253. CONCLUSIONS: The optimal therapeutic synergy was achieved when irinotecan was administered 24 h before 5-Flurouracil. Sensitivity to this combination was associated with poor differentiation status, higher cyclin A index, recruitment of cells into S phase, and induction of Bax expression and apoptosis.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Carcinoma, Squamous Cell/pathology , Fluorouracil/therapeutic use , Head and Neck Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Cell Cycle , Cell Line, Tumor , Female , Head and Neck Neoplasms/drug therapy , Humans , Immunohistochemistry , Irinotecan , Kinetics , Maximum Tolerated Dose , Mice , Mice, Nude , Neoplasm Transplantation , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
15.
Biochem Biophys Res Commun ; 295(2): 435-44, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12150968

ABSTRACT

A novel karenitecin, BNP1350, is a topoisomerase I-targeting anticancer agent with significant antitumor activity in vitro and in vivo. A BNP1350-resistant human head and neck carcinoma A253 cell line, denoted A253/BNPR, was developed. The A253/BNPR cell line was approximately 9-fold resistant to BNP1350 and 4-fold cross-resistant to another topoisomerase I inhibitor SN-38, the active metabolite of irinotecan. After drug treatment with equimolar concentrations of BNP1350 (0.7 microM) for 2h, activation of the DNA double-strand break repair protein complexes was similar in the two cell lines, suggesting that DNA dsb repair is not attributable to resistance to BNP1350 in the A253/BNPR cells. Cell cycle analysis indicates that the A253 cell line accumulated primarily in S phase, but G(2) phase accumulation was observed in the A253/BNPR cell line at 48 h after drug removal. Elevated chk1 phosphorylation at Ser(345) following DNA damage induced by BNP1350 was accompanied by G(2) accumulation in the A253/BNPR cell line, while exposure to equimolar concentrations of BNP1350 (0.7 microM) induced S-phase arrest and no increased phosphorylation of chk1 at Ser(345) in the A253 cell line. Under the same conditions, increased chk1 activity was observed in the A253/BNPR cell line, but not in the A253 cell line. Moreover, stimulated binding of 14-3-3 proteins to chk1 was observed in BNP1350-treated A253/BNPR cells. To confirm relationship between chk1 expression/phosphorylation and drug resistance to topo I poisons, we examined the effects of chk1 or chk2 antisense oligonucleotides on the cellular growth inhibition. Chk1 antisense oligonucleotide can sensitize the A253/BNPR cells to killing by topo I inhibitor BNP1350, but no significant sensitization of BNP1350-induced growth inhibition was observed in the drug-sensitive cell line. Chk2 antisense oligonucleotide has only a small sensitization effect on BNP1350-induced growth inhibition in both cell lines. The data indicate that the chk1 signaling pathways that mediate cell cycle checkpoint are associated with cellular resistance to BNP1350 in the A253/BNPR cell line.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , G2 Phase , Mitosis , Protein Kinases/metabolism , Signal Transduction , Topoisomerase I Inhibitors , 14-3-3 Proteins , Cell Division/drug effects , Checkpoint Kinase 1 , DNA Damage , DNA Repair , Humans , Oligonucleotides, Antisense/metabolism , Phosphorylation , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...