Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Sci Rep ; 11(1): 22500, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795360

ABSTRACT

Mice have served as an excellent model to understand the etiology of lung cancer for years. However, data regarding dual-stage carcinogenesis of lung squamous cell carcinoma (SCC) remain elusive. Therefore, we aim to develop pre-malignant (PM) and malignant (M) lung SCC in vivo using N-nitroso-tris-chloroethylurea (NTCU). BALB/C mice were allotted into two main groups; PM and M groups which received treatment for 15 and 30 weeks, respectively. Then, the mice in each main group were allotted into three groups; control, vehicle, and cancer (n = 6), which received normal saline, 70% acetone, and 0.04 M NTCU by skin painting, respectively. Histopathologically, we discovered a mix of hyperplasia, metaplasia, and dysplasia lesions in the PM group and intracellular bridge; an SCC feature in the M group. The M group was positive for cytokeratin 5/6 protein which confirmed the lung SCC subtype. We also found significantly higher (P < 0.05) epithelium thickness in the cancer groups as compared to the vehicle and control groups at both the PM and M. Overall, this study discovered that NTCU is capable of developing PM and M lung SCC in mice model at appropriate weeks and the vehicle group was suggested to be adequate as control group for future research.


Subject(s)
Carcinoma, Non-Small-Cell Lung/physiopathology , Carcinoma, Squamous Cell/physiopathology , Carmustine/analogs & derivatives , Lung Neoplasms/physiopathology , Animals , Biomarkers, Tumor/metabolism , Carcinogens , Carcinoma, Non-Small-Cell Lung/chemically induced , Carcinoma, Squamous Cell/chemically induced , Disease Models, Animal , Epithelium/metabolism , Female , Immunohistochemistry , Keratins/metabolism , Lung Neoplasms/chemically induced , Mice , Mice, Inbred BALB C
2.
Cancer Prev Res (Phila) ; 14(3): 307-312, 2021 03.
Article in English | MEDLINE | ID: mdl-33115782

ABSTRACT

Tobacco smoke-induced squamous cell lung cancer (SCC) develops from endobronchial dysplastic lesions that progress to invasive disease. A reproducible murine model recapitulating histologic progression observed in current and former smokers will advance testing of new preventive and therapeutic strategies. Previous studies show that prolonged topical application of N-nitroso-tris-chloroethylurea (NTCU) generates a range of airway lesions in sensitive mice similar to those induced by chronic tobacco smoke exposure in humans. To improve the current NTCU model and better align it with human disease, NTCU was applied to mice twice weekly for 4-5 weeks followed by a recovery period before cigarette smoke (CS) or ambient air (control) exposure for an additional 3-6 weeks. Despite the short time course, the addition of CS led to significantly more premalignant lesions (PML; 2.6 vs. 0.5; P < 0.02) and resulted in fewer alveolar macrophages (52,000 macrophages/mL BALF vs. 68,000; P < 0.05) compared with control mice. This improved NTCU + CS model is the first murine SCC model to incorporate tobacco smoke and is more amenable to preclinical studies because of the increased number of PML, decreased number of mice required, and reduced time needed for PML development.


Subject(s)
Carcinoma, Squamous Cell/pathology , Carmustine/analogs & derivatives , Disease Models, Animal , Precancerous Conditions/pathology , Respiratory System/pathology , Smoke/adverse effects , Tobacco Smoke Pollution/adverse effects , Animals , Carcinoma, Squamous Cell/chemically induced , Carmustine/toxicity , Female , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred A , Precancerous Conditions/chemically induced , Respiratory System/drug effects
3.
Histol Histopathol ; 35(10): 1159-1170, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32893871

ABSTRACT

BACKGROUND: Lung cancer is the leading cause of cancer-related deaths, and squamous cell carcinoma (SCC) is one of the most common types of lung cancer. Chemoprevention of lung cancer has gained increasing popularity as an alternative to treatment in reducing the burden of lung cancer. Pterostilbene (PS) may be developed as a chemopreventive agent due to its pharmacological activities, such as anti-proliferative, anti-inflammatory and antioxidant properties. This study aimed to investigate the effect of PS on the development of lung SCC in the mouse model. METHODS: A total of 24 seven-week-old female Balb/C mice were randomly categorised into four groups, including two control groups comprising the N-nitroso-trischloroethylurea (NTCU)-induced lung SCC and vehicle control (VC) groups and two treatment groups comprising the 10mg/kg PS (PS10) and 50mg/kg PS (PS50) groups. All lung organs were harvested at week 26 for histopathological analysis. RESULTS: All PS treatment groups showed chemopreventive activity by inhibiting the progression of lung SCC formation with PS10, resulting in mild hyperplasia, and PS50 was completely reversed in the normal bronchial epithelium layer compared with the VC group. PS treatment also reduced the expression of cytokeratin 5/6 in the bronchial epithelium layer. Both PS10 and PS50 significantly reduced the epithelium thickness compared to the NTCU group (p<0.05). PS is a potential chemopreventive agent against lung SCC growth by suppressing the progression of pre-malignant lesions and reducing the thickness of the bronchial epithelium. CONCLUSIONS: The underlying molecular mechanisms of PS in lung SCC should be further studied.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Squamous Cell/prevention & control , Lung Neoplasms/prevention & control , Lung/drug effects , Stilbenes/pharmacology , Animals , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carmustine/analogs & derivatives , Disease Models, Animal , Female , Keratin-15/metabolism , Keratin-6/metabolism , Lung/metabolism , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred BALB C
4.
Cancer Prev Res (Phila) ; 12(8): 507-516, 2019 08.
Article in English | MEDLINE | ID: mdl-31101634

ABSTRACT

In order to translate new treatments to the clinic, it is necessary to use animal models that closely recapitulate human disease. Lung cancer develops after extended exposure to carcinogens. It has one of the highest mutation rates of all cancer and is highly heterogenic. Topical treatment with N-nitrosotris-(2-chloroethyl)urea (NTCU) induces lung squamous cell carcinoma (SCC) with nonsynonymous mutation rates similar to those reported for human non-small cell lung cancer. However, NTCU induces lung cancer with variable efficacy and toxicity depending on the mouse strain. A detailed characterization of the NTCU model is needed. We have compared the effect of three different NTCU doses (20, 30, and 40 mmol/L) in female and male of NIH Swiss, Black Swiss, and FVB mice on tumor incidence, survival, and toxicity. The main findings in this study are (1) NIH Swiss mice present with a higher incidence of SCC and lower mortality compared with Black Swiss and FVB mice; (2) 30 mmol/L NTCU dose induces SCC at the same rate and incidence as the 40 mmol/L dose with lower mortality; (3) female mice present higher grade and incidence of preinvasive lesions and SCC compared with males; (4) NTCU-induced transformation is principally within the respiratory system; and (5) NTCU treatment does not affect the ability to elicit a specific adaptive immune response. This study provides a reference point for experimental designs to evaluate either preventive or therapeutic treatments for lung SCC, including immunotherapies, before initiating human clinical trials.


Subject(s)
Carcinogens/toxicity , Carcinoma, Squamous Cell/epidemiology , Lung Neoplasms/epidemiology , Lung/pathology , Animals , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Carmustine/analogs & derivatives , Carmustine/toxicity , Cell Transformation, Neoplastic/chemically induced , Dose-Response Relationship, Drug , Female , Incidence , Lung/drug effects , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Male , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/epidemiology , Neoplasms, Experimental/pathology , Risk Factors , Sex Factors
5.
Cancer Sci ; 107(2): 123-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663681

ABSTRACT

The role of cells expressing stem cell markers deltaNp63 and CD44v has not yet been elucidated in peripheral-type lung squamous cell carcinoma (pLSCC) carcinogenesis. Female A/J mice were painted topically with N-nitroso-tris-chloroethylurea (NTCU) for induction of pLSCC, and the histopathological and molecular characteristics of NTCU-induced lung lesions were examined. Histopathologically, we found atypical bronchiolar hyperplasia, squamous metaplasia, squamous dysplasia, and pLSCCs in the treated mice. Furthermore, we identified deltaNp63(pos)CD44v(pos)CK5/6(pos)CC10(pos) clara cells as key constituents of early precancerous atypical bronchiolar hyperplasia. In addition, deltaNp63(pos)CD44v(pos) cells existed throughout the atypical bronchiolar hyperplasias, squamous metaplasias, squamous dysplasias, and pLSCCs. Overall, our findings suggest that NTCU induces pLSCC through an atypical bronchiolar hyperplasia-metaplasia-dysplasia-SCC sequence in mouse lung bronchioles. Notably, Ki67-positive deltaNp63(pos)CD44v(pos) cancer cells, cancer cells overexpressing phosphorylated epidermal growth factor receptor and signal transducer and activator of transcription 3, and tumor-associated macrophages were all present in far greater numbers in the peripheral area of the pLSCCs compared with the central area. These findings suggest that deltaNp63(pos)CD44v(pos) clara cells in mouse lung bronchioles might be the origin of the NTCU-induced pLSCCs. Our findings also suggest that tumor-associated macrophages may contribute to creating a tumor microenvironment in the peripheral area of pLSCCs that allows deltaNp63(pos)CD44v(pos) cancer cell expansion through activation of epidermal growth factor receptor signaling, and that exerts an immunosuppressive effect through activation of signal transducer and activator of transcription 3 signaling.


Subject(s)
Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Precancerous Conditions/pathology , Tumor Microenvironment/immunology , Animals , Carcinogens/toxicity , Carcinoma, Squamous Cell/immunology , Carmustine/analogs & derivatives , Carmustine/toxicity , Disease Models, Animal , Disease Progression , Female , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/immunology , Immune Tolerance/immunology , Immunohistochemistry , Lung Neoplasms/immunology , Macrophages/pathology , Mice , Phosphoproteins/biosynthesis , Phosphoproteins/immunology , Trans-Activators/biosynthesis , Trans-Activators/immunology , Tumor Escape/immunology
6.
PLoS One ; 10(4): e0122823, 2015.
Article in English | MEDLINE | ID: mdl-25860262

ABSTRACT

Squamous cell lung cancer (SCC) is the second leading cause of lung cancer death in the US and has a 5-year survival rate of only 16%. Histological changes in the bronchial epithelium termed dysplasia are precursors to invasive SCC. However, the cellular mechanisms that cause dysplasia are unknown. To fill this knowledge gap, we used topical application of N-nitroso-tris chloroethylurea (NTCU) for 32 weeks to induce squamous dysplasia and SCC in mice. At 32 weeks the predominant cell type in the dysplastic airways was Keratin (K) 5 and K14 expressing basal cells. Notably, basal cells are extremely rare in the normal mouse bronchial epithelium but are abundant in the trachea. We therefore evaluated time-dependent changes in tracheal and bronchial histopathology after NTCU exposure (4, 8, 12, 16, 25 and 32 weeks). We show that tracheal dysplasia occurs significantly earlier than that of the bronchial epithelium (12 weeks vs. 25 weeks). This was associated with increased numbers of K5+/K14+ tracheal basal cells and a complete loss of secretory (Club cell secretory protein expressing CCSP+) and ciliated cells. TUNEL staining of NTCU treated tissues confirmed that the loss of CCSP+ and ciliated cells was not due to apoptosis. However, mitotic index (measured by bromodeoxyuridine incorporation) showed that NTCU treatment increased proliferation of K5+ basal cells in the trachea, and altered bronchial mitotic population from CCSP+ to K5+ basal cells. Thus, we demonstrate that NTCU-induced lung epithelial dysplasia starts in the tracheal epithelium, and is followed by basal cell metaplasia of the bronchial epithelium. This analysis extends our knowledge of the NTCU-SCC model by defining the early changes in epithelial cell phenotypes in distinct airway locations, and this may assist in identifying new targets for future chemoprevention studies.


Subject(s)
Bronchi/pathology , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Carmustine/analogs & derivatives , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Trachea/pathology , Animals , Biomarkers, Tumor , Carmustine/adverse effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Mice , Mitotic Index , Neoplasm Grading , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
7.
Cancer Prev Res (Phila) ; 8(1): 77-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25403850

ABSTRACT

Inflammatory conditions of the lung such as chronic obstructive pulmonary disease (COPD) are known to increase lung cancer risk, particularly lung squamous cell carcinoma (LSCC). In the present study, we developed a mouse model of inflammation-driven LSCC that was induced by N-nitroso-trischloroethylurea (NTCU) and enhanced by lipopolysaccharide (LPS), a potent proinflammatory agent contained in tobacco and tobacco smoke, and determined the chemopreventive effects of BioResponse diindolylmethane (DIM) in the same model. Compared with mice treated with NTCU alone, mice treated with the combination of NTCU and LPS had a 9-fold increase in the number of bronchioles with LSCC. Also, compared with mice treated with LPS alone, mice treated with NTCU plus LPS showed significantly increased expression of the inflammatory cytokines IL1α, IL6, and TNFα (all three increased about 7-fold). Parallel to the increased cytokine gene expression, the NTCU plus LPS-treated group exhibited significantly enhanced activation of NF-κB, STAT3, ERK, p-38, and Akt, expression of p53, COX-2, and Mcl-1, and NF-κB- and STAT3-DNA binding in the lung. Dietary administration of DIM (10 µmol/g diet or 2,460 ppm) to mice treated with NTCU plus LPS reduced the incidence of LSCC by 2-fold, suppressed activation/expression of proinflammatory and procarcinogenic proteins and NF-κB- and STAT3-DNA binding, but not the expression of cytokines and p53. This study highlights the potential significance of our mouse model to identify promising drugs or dietary agents for the chemoprevention of human LSCC and that DIM is a very good candidate for clinical lung cancer chemoprevention trials.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Indoles/administration & dosage , Inflammation/drug therapy , Lung Neoplasms/drug therapy , Animals , Carmustine/analogs & derivatives , Carmustine/chemistry , Cytokines/metabolism , Diet , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Indoles/chemistry , Lipopolysaccharides/chemistry , Lung/metabolism , Lung Neoplasms/metabolism , Mice , NF-kappa B/metabolism
8.
Cancer Prev Res (Phila) ; 7(11): 1149-59, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25245764

ABSTRACT

Honokiol is an important bioactive compound found in the bark of Magnolia tree. It is a nonadipogenic PPARγ agonist and capable of inhibiting the growth of a variety of tumor types both in vitro and in xenograft models. However, to fully appreciate the potential chemopreventive activity of honokiol, a less artificial model system is required. To that end, this study examined the chemopreventive efficacy of honokiol in an initiation model of lung squamous cell carcinoma (SCC). This model system uses the carcinogen N-nitroso-trischloroethylurea (NTCU), which is applied topically, reliably triggering the development of SCC within 24 to 26 weeks. Administration of honokiol significantly reduced the percentage of bronchial that exhibit abnormal lung SCC histology from 24.4% bronchial in control to 11.0% bronchial in honokiol-treated group (P = 0.01) while protecting normal bronchial histology (present in 20.5% of bronchial in control group and 38.5% of bronchial in honokiol-treated group. P = 0.004). P63 staining at the SCC site confirmed the lung SCCs phenotype. In vitro studies revealed that honokiol inhibited lung SCC cells proliferation, arrested cells at the G1-S cell-cycle checkpoint, while also leading to increased apoptosis. Our study showed that interfering with mitochondrial respiration is a novel mechanism by which honokiol changed redox status in the mitochondria, triggered apoptosis, and finally leads to the inhibition of lung SCC. This novel mechanism of targeting mitochondrial suggests honokiol as a potential lung SCC chemopreventive agent.


Subject(s)
Anticarcinogenic Agents/pharmacology , Biphenyl Compounds/pharmacology , Lignans/pharmacology , Lung Neoplasms/pathology , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Bronchi/drug effects , Carcinogens/chemistry , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carmustine/analogs & derivatives , Carmustine/chemistry , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/drug therapy , Mice , Mitochondria/drug effects , Neoplasm Transplantation , Oxidation-Reduction , Reactive Oxygen Species/metabolism
9.
Cancer Sci ; 104(12): 1560-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24106881

ABSTRACT

Targeted treatments for lung cancer based on pathological diagnoses are required to enhance therapeutic efficacy. There are few well-established animal models for lung squamous cell carcinoma although several highly reproducible mouse models for lung adenoma and adenocarcinoma are available. This study was carried out to establish a new lung squamous cell carcinoma mouse model. In the first experiment, female A/J mice were painted topically on back skin twice weekly with 75 µL 0.013 M N-nitroso-tris-chloroethylurea for 2, 4, and 8 weeks (n = 15-20 per group) as initiation of lung lesions, and surviving mice were killed at 18 weeks. In the second experiment, mice were treated as above for 4 weeks and killed at 6, 12, or 18 weeks (n = 3 per group). Lung lobes were subjected to histopathological, immunohistochemical, immunoblotting, and ultrastructural analyses. In the case of treatment for 2, 4, and 8 weeks, incidences of lung squamous cell carcinoma were 25, 54, and 71%, respectively. Cytokeratin 5/6 and epidermal growth factor receptor were clearly expressed in dysplasia and squamous cell carcinoma. Desmosomes and tonofilaments developed in the squamous cell carcinoma. Considering the carcinogenesis model, we conclude that 2 or 4 weeks of N-nitroso-tris-chloroethylurea treatment may be suitable for investigating new chemicals for promotional or suppressive effects on lung squamous cell carcinoma.


Subject(s)
Carcinogenesis/chemically induced , Carcinoma, Squamous Cell/pathology , Carmustine/analogs & derivatives , Disease Models, Animal , Lung Neoplasms/pathology , Mice , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/mortality , Carmustine/toxicity , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P450 Family 2 , ErbB Receptors/biosynthesis , Female , Lung/drug effects , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/mortality , Mice, Inbred A , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Survival Rate
10.
Acta Crystallogr C ; 68(Pt 3): o144-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22382550

ABSTRACT

Nimustine hydrochloride [systematic name: 4-amino-5-({[N-(2-chloroethyl)-N-nitrosocarbamoyl]amino}methyl)-2-methylpyrimidin-1-ium chloride], C(9)H(14)ClN(6)O(2)(+)·Cl(-), is a prodrug of CENU (chloroethylnitrosourea) and is used as a cytostatic agent in cancer therapy. Its crystal structure was determined from laboratory X-ray powder diffraction data. The protonation at an N atom of the pyrimidine ring was established by solid-state NMR spectroscopy.


Subject(s)
Carmustine/analogs & derivatives , Crystallography, X-Ray , Carmustine/chemistry , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Models, Molecular , Nimustine , Powders , X-Ray Diffraction
11.
Cancer Prev Res (Phila) ; 5(2): 283-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086679

ABSTRACT

Squamous cell carcinoma (SCC) and premalignant endobronchial lesions have been difficult to study in murine models. In this study, we evaluate the topical N-nitroso-tris-chloroethylurea (NTCU) murine SCC model, determine the extent to which resulting premalignant airway dysplasia develops, discuss clinicopathologic grading criteria in lesion progression, and confirm that immunohistochemical (IHC) staining patterns are consistent with those observed in human endobronchial dysplasia and SCC. Male and female FVB mice were treated biweekly with topical NTCU (4, 8, or 40 mmol/L) or vehicle for 32 weeks. Following sacrifice, squamous cell lesions were enumerated and categorized into the following groups: flat atypia, low-grade dysplasia, high-grade dysplasia, and invasive SCC. The 40 mmol/L NTCU concentration produced the entire spectrum of premalignant dysplasias and squamous cell carcinomas, but was associated with poor survival. Concentrations of 4 and 8 mmol/L NTCU were better tolerated and produced only significant levels of flat atypia. Squamous origin of the range of observed lesions was confirmed with IHC staining for cytokeratin 5/6, p63, thyroid transcription factor-1 (TTF-1), and Napsin-A. This study shows that topical application of high-dose NTCU produces endobronchial premalignant lesions with classic squamous characteristics and should allow for improved preclinical evaluation of potential chemopreventive agents.


Subject(s)
Bronchial Neoplasms/chemically induced , Carcinogens/toxicity , Carcinoma, Squamous Cell/chemically induced , Carmustine/analogs & derivatives , Disease Models, Animal , Precancerous Conditions/chemically induced , Administration, Topical , Animals , Bronchial Neoplasms/mortality , Bronchial Neoplasms/pathology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carmustine/toxicity , Female , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Precancerous Conditions/mortality , Precancerous Conditions/pathology , Survival Rate
12.
Invest New Drugs ; 24(4): 347-51, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16502354

ABSTRACT

PURPOSE: To evaluate the activity and toxicity of SarCNU, an oral chloroethylnitrosourea in patients with recurrent or metastatic colorectal cancer who have progressed after first-line chemotherapy. PATIENTS AND METHODS: Eighteen patients with recurrent or metastatic colorectal cancer following first-line chemotherapy were treated with SarCNU 860 mg/m2 orally day 1, 5 and 9 every 6 weeks. The patient's median age was 64 and the ECOG performance status was 0 in six, 1 in eleven and 2 in one patients. All patients were evaluable for toxicity and 16 were evaluable for response. RESULTS: There were no objective responses (0%). One patient had stable disease and 15 had progressive disease at their first follow-up assessment. Median survival was 7.36 months (3.75-7.49 95% C.I.). Neutropenia and thrombocytopenia were the most severe toxicities (grade 3-4 in six and nine patients respectively). Pulmonary toxicity was also seen in five patients who had a drop of DLCO grade from baseline and two patients who had a fall in FVC from baseline. CONCLUSIONS: SarCNU is inactive in recurrent or metastatic colorectal patients who have progressed after first-line chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carmustine/analogs & derivatives , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Canada , Carmustine/administration & dosage , Carmustine/adverse effects , Carmustine/therapeutic use , Colorectal Neoplasms/prevention & control , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasm Metastasis/drug therapy , Survival Rate
13.
Int J Oncol ; 27(4): 1131-40, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142332

ABSTRACT

2-Chloroethyl-3-sarcosinamide-1-nitrosourea (SarCNU) has been used to treat patients with advanced solid tumours. However, the molecular mechanisms are not well understood. In the present study, we report that SarCNU inhibited proliferation of human HK-1 and CNE-2 nasopharyngeal carcinoma (NPC) in vivo and in vitro. In vitro study showed that wild-type p53 HK-1 cells were 3-fold more sensitive to SarCNU than p53 mutant CNE-2 cells. G2/M arrest, reduction in p21(Cip1/Waf1) and inactivation of cellular cdc-2 activity were seen in both SarCNU-treated HK-1 and CNE-2 cells. Upregulation of p53, phosphorylated p53 at Ser15 and biochemical markers for apoptosis, such as cleaved caspase-3, cleaved caspase-7 and cleaved PARP, were observed in SarCNU-treated HK-1 but not CNE-2 cells. The levels of cyclin B1, Wee1 and phosphorylated cdc-2 but not total cdc-2 in HK-1 cells were significantly reduced by SarCNU treatment. In contrast to HK-1 cells, decrease in total cdc-2 but increase in phosphorylated cdc-2 at Tyr15, cyclin B1 and Wee1 was observed in CNE-2 cells treated with SarCNU. Introduction of mutant p53 into HK-1 cells resulted in growth enhancement in vivo and increased resistance to SarCNU-induced apoptosis in vitro. Furthermore, CNE-2 cells transfected with wild-type p53 became susceptible to SarCNU-induced apoptosis in vitro but not their growth rate in vivo. The data indicate that in NPC cells SarCNU-induced apoptosis was p53-dependent while SarCNU-induced G2/M arrest was mediated by altering the levels of cyclin B1-cdc-2 complex and phosphorylation of cdc-2 at Tyr15 resulting in inactivation of cellular cdc-2 activity. Our data suggest a potential use of SarCNU in the treatment of NPC.


Subject(s)
Carcinoma/pathology , Carmustine/analogs & derivatives , Nasopharyngeal Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , CDC2 Protein Kinase/metabolism , Carmustine/pharmacology , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Division , Cell Proliferation , Cell Survival , Cyclin B/metabolism , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , G2 Phase , Genes, p53 , Genetic Therapy/methods , Humans , Immunoprecipitation , In Vitro Techniques , Male , Mice , Mice, SCID , Mitosis , Mutation , Nuclear Proteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Time Factors , Transfection , Tyrosine/chemistry , Up-Regulation
14.
Invest New Drugs ; 23(6): 591-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16034522

ABSTRACT

PURPOSE: A multi-centre phase II study of SarCNU-a novel chloroethylnitrosourea (CNU)-in patients with recurrent malignant glioma to assess response rate, survival and effects of treatment. PATIENTS AND METHODS: Ten patients with histologically proven malignant glioma (seven with glioblastoma multiforme (GBM) and three with anaplastic astrocytoma) received SarCNU (860 mg/m(2)) orally on days 1, 5 and 9 on a 6 week schedule. RESULTS: A total of ten patients were treated on protocol before accrual was suspended for a high rate of pulmonary toxicity. Of eight evaluable patients, five demonstrated at least one grade deterioration in DLCO from baseline. This necessitated premature closure of the trial. Stable disease was seen in five of seven evaluable patients (median duration 4.8 months; range 0.8-9.2) with progressive disease in the remainder. CONCLUSION: Despite promising preclinical data, SarCNU caused pulmonary toxicity in patients with recurrent malignant glioma and we plan no further studies in this indication.


Subject(s)
Antineoplastic Agents/therapeutic use , Astrocytoma/drug therapy , Carmustine/analogs & derivatives , Central Nervous System Neoplasms/drug therapy , Glioblastoma/drug therapy , Lung/drug effects , Academies and Institutes , Adult , Aged , Antineoplastic Agents/adverse effects , Canada , Carbon Monoxide/metabolism , Carmustine/adverse effects , Carmustine/therapeutic use , Female , Humans , Lung/physiopathology , Male , Middle Aged , Neoplasm Recurrence, Local , Pulmonary Diffusing Capacity/drug effects
15.
J Cell Physiol ; 204(3): 785-91, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15754328

ABSTRACT

Hepatocellular carcinoma (HCC) is a major health problem in the Asia-Pacific region, with high incidence and mortality rate. There is currently no effective treatment for inoperable cases that represent the vast majority of patients. In the present study, we report that in vitro treatment of primary hepatoma, HepG2 (wild-type p53), PLC/PRF/5 (p53-mutant), and Hep3B (p53-deleted) cells with 2-chloroethyl-3-sarcosinamide-1-nitrosourea (SarCNU) resulted in upregulation of p53, p21(Cip1/Waf1), phosphorylated cdc-2 at Tyr15 in wild-type p53 cells and phosphorylation of cdc-2 at Tyr15 in p53-mutant or p53-deleted hepatoma cells. This was accompanied by the reduction in cdc-2 kinase activity and G(2)/M cell cycle arrest. These findings indicate that SarCNU-induced G(2)/M growth arrest in hepatoma cells by a p53-independent phosphorylation of cdc-2. Our data suggest the potential use of SarCNU in treatment of HCC.


Subject(s)
CDC2 Protein Kinase/metabolism , Carmustine/analogs & derivatives , G2 Phase/drug effects , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mitosis/drug effects , Phosphotyrosine/metabolism , Carmustine/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Humans , Liver Neoplasms/genetics , Phosphorylation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Cancer ; 101(12): 2881-91, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15529313

ABSTRACT

BACKGROUND: Prostate carcinoma is the most commonly occurring malignancy in men. Although 2-chloroethyl-3-sarcosinamide-1-nitrosourea (SarCNU), an analog of the chloroethylnitrosoureas, has been used in the treatment of advanced solid tumors, the molecular mechanisms underlying the antineoplastic activity of this agent are not well understood. In the current study, the authors sought to investigate the effects of SarCNU on prostate carcinoma cell growth in vivo and in vitro. METHODS: Male SCID mice underwent subcutaneous implantation (on both flanks) of human CWR-22 and CWR-22R prostate carcinoma xenografts. Mice were treated with either vehicle or 60 or 80 mg SarCNU per kg body weight for 5 days, with tumor growth being assessed every 3 days. Animals were sacrificed 21 days after the final injection, and tumors subsequently were collected, weighed, and processed for analysis. Immunohistochemical analyses were performed to obtain data on the localization of p53 and p21Cip1/Waf1. Cell counting, terminal deoxynucleotidyltransferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) assays, cell cycle analyses, Western blotting, and in vitro kinase assays were performed to determine the effects of SarCNU on growth, apoptosis, cell cycle arrest, cell cycle-regulated protein levels, and Cdc-2 activity, respectively. RESULTS: SarCNU reduced tumor incidence and inhibited the growth of CWR-22 and CWR-22R xenografts. In addition, treatment with this agent led to increases in p21Cip1/Waf1 levels and p53 phosphorylation at Ser15. In vitro administration of SarCNU to cells with wild-type p53 (LNCaP and primary CWR-22 cells) and cells with mutant p53 (PC-3 cells) resulted in G2/M arrest and the reduction of cellular Cdc-2 activity. Up-regulation of p53 levels, p53 phosphorylation at Ser15, and p21Cip1/Waf1 levels in primary CWR-22 and LNCaP cells, as well as up-regulation of Cdc-2 phosphorylation at Tyr15 in PC-3 cells, was detected. CONCLUSIONS: SarCNU induced G2/M arrest in prostate carcinoma cells via p53-dependent up-regulation of p21Cip1/Waf1 and p53-independent phosphorylation of Cdc-2 at Tyr15. These findings suggest a potential role for SarCNU in the treatment of prostate malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/pathology , Carmustine/analogs & derivatives , Carmustine/pharmacology , Genes, p53 , Prostatic Neoplasms/pathology , Animals , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, SCID , Neoplasm Transplantation , Phosphorylation , Prostatic Neoplasms/prevention & control , Tumor Cells, Cultured
17.
Mol Cancer Ther ; 3(7): 849-60, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15252146

ABSTRACT

We identified five structurally related dimethane sulfonates with putative selective cytotoxicity in renal cancer cell lines. These compounds have a hydrophobic moiety linked to a predicted alkylating group. A COMPARE analysis with the National Cancer Institute Anticancer Drug Screen standard agent database found significant correlations between the IC50 of the test compounds and the IC50 of alkylating agents (e.g., r = 0.68, P < 0.00001 for chlorambucil). In this report, we examined whether these compounds had activities similar to those of conventional alkylating agents. In cytotoxicity studies, chlorambucil-resistant Walker rat carcinoma cells were 4- to 11-fold cross-resistant to the test compounds compared with 14-fold resistant to chlorambucil. To determine effects on cell cycle progression, renal cell carcinoma (RCC) line 109 was labeled with bromodeoxyuridine prior to drug treatment. Complete cell cycle arrest occurred in cells treated with an IC90 dose of NSC 268965. p53 protein levels increased as much as 5.7-fold in RCC line 109 and as much as 20.4-fold in breast cancer line MCF-7 following an 18-hour drug exposure. Finally, DNA-protein cross-links were found following a 6-hour pretreatment with all compounds. Thus, the dimethane sulfonate analogues have properties expected of some alkylating agents but, unlike conventional alkylating agents, appear to possess activity against RCC.


Subject(s)
Alkylating Agents/chemistry , Alkylating Agents/toxicity , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Mesylates/chemistry , Mesylates/toxicity , Alkylating Agents/therapeutic use , Animals , Bromodeoxyuridine/analysis , Busulfan/analogs & derivatives , Carmustine/analogs & derivatives , Cell Cycle/drug effects , DNA Damage , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Humans , Inhibitory Concentration 50 , Mesylates/therapeutic use , Rats , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism , Yeasts/drug effects
18.
Blood Cells Mol Dis ; 31(1): 48-50, 2003.
Article in English | MEDLINE | ID: mdl-12850483

ABSTRACT

The therapeutic combination O(6)-benzylguanine (BG) and 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) can be used to select for P140K methylguanine-DNA-methyltransferase (P140K MGMT) transduced hematopoietic progenitors both in vitro and in vivo. SarCNU is a new alkylating agent which has advantages over BCNU. We tested the ability of P140K MGMT transduced human CD34(+) cells to resist SarCNU treatment and be selected for in vitro. BG/SarCNU exposure led to an increase in the proportion of transduced cells from 13 to 27%. The IC(90) was increased sixfold by P140K MGMT transduction. These results suggest that SarCNU may be a suitable agent for in vivo selection strategies.


Subject(s)
Carmustine/analogs & derivatives , Carmustine/pharmacology , Cell Separation/methods , Hematopoietic Stem Cells/metabolism , O(6)-Methylguanine-DNA Methyltransferase/genetics , Transduction, Genetic/methods , Antigens, CD34 , Antineoplastic Agents/pharmacology , Cell Survival , Drug Resistance, Neoplasm , Hematopoietic Stem Cells/drug effects , Humans , Mutation, Missense
19.
J Clin Oncol ; 21(2): 232-40, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12525514

ABSTRACT

PURPOSE: 2-Chloroethyl-3-sarcosinamide-1-nitrosourea (SarCNU) is a novel chloroethylnitrosourea that demonstrates selective cytotoxicity in athymic mice bearing human glioma. SarCNU demonstrates selective cytotoxicity in vitro against human glioma at least in part because of the selective SarCNU uptake by the extraneuronal monoamine transporter. The purpose of this phase I study was to determine the maximum-tolerated dose (MTD), the toxicity profile, the pharmacokinetics profile, and recommended phase II dose. PATIENTS AND METHODS: Forty-three eligible patients with advanced solid tumors were enrolled. SarCNU was administered orally on days 1,5, and 9 every 28 days. The dose ranged from 30 to 1,075 mg/m2. Pharmacokinetic evaluation was done on the first cycle (one dose was given intravenously on day 1 or 5 of the first cycle to determine bioavailability). RESULTS: Delayed myelosuppression (thrombocytopenia and neutropenia occurring 4 to 6 weeks after administration) was the dose-limiting toxicity (DLT). Anemia occurred but was mild. Nonhematologic toxicity was generally mild, but one patient died with pulmonary toxicity that was probably secondary to SarCNU. There were no partial or complete responses, but eight patients had stable disease for 19 to 46 weeks. The oral bioavailability of SarCNU was 80% +/- 37%. The terminal phase half-life was similar after intravenous (58.4 +/- 23.5 minutes) or oral (64.0 +/- 34.8 minutes) administration. The total plasma clearance was 20.4 +/- 8.8 L/h/m2, and the apparent volume of distribution was 29.9 +/- 17.6 L/m2. The area under the plasma concentration-time profile increased proportionally with the dose, and the pharmacokinetics seemed to be independent of the route of administration and the number of doses. CONCLUSION: SarCNU was well tolerated and the MTD was 1,075 mg/m2. The recommended starting dose for phase II trials is 860 mg/m2 orally on days 1, 5, and 9 every 6 weeks.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carmustine/analogs & derivatives , Carmustine/pharmacokinetics , Neoplasms/metabolism , Administration, Oral , Adult , Aged , Biological Availability , Female , Half-Life , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Middle Aged , Thrombocytopenia/chemically induced
20.
Int J Pharm ; 249(1-2): 257-64, 2002 Dec 05.
Article in English | MEDLINE | ID: mdl-12433453

ABSTRACT

The stability of SarCNU (NSC364432), 1-(2-chloroethyl)-3-sarcosinamide-1-nitrosourea in several pharmaceutically acceptable solvents was investigated by high pressure liquid chromatography (HPLC). The influences of light, ionic strength, pH, buffer concentration, and the following excipients: benzyl alcohol, ascorbic acid, sodium bisulfite, and disodium EDTA were studied at room temperature. The stability of the drug was also determined in water, EtOH, PG, Capmul PG, DMSO, and in different combinations of these cosolvents at four different temperatures. The degradation of the drug, which is catalyzed not only by general but also by specific acid and base, follows first order kinetics. Antioxidants, EDTA, and light have no effect on the degradation rate, suggesting oxidation is not a major degradation pathway. The t(90) in pure cosolvent is 25-50 times higher than that in water or semi-aqueous vehicles. Neat EtOH can be used to store the drug in a nonaqueous concentrate that is diluted with aqueous solvent prior to injection.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Carmustine/analogs & derivatives , Carmustine/chemistry , Carmustine/pharmacokinetics , Chemistry, Pharmaceutical , Drug Stability
SELECTION OF CITATIONS
SEARCH DETAIL
...