Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Mol Cancer ; 21(1): 193, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36203195

ABSTRACT

EGFR-TKI targeted therapy is one of the most effective treatments for lung cancer patients harboring EGFR activating mutations. However, inhibition response is easily attenuated by drug resistance, which is mainly due to bypass activation or downstream activation. Herein, we established osimertinib-resistant cells by stepwise dose-escalation in vitro and an osimertinib-resistant patient-derived xenograft model through persistent treatment in vivo. Phosphorylated proteomics identified that MEK1 and AKT1/2 were abnormally activated in resistant cells compared with parental cells. Likewise, EGFR inhibition by osimertinib induced activation of MEK1 and AKT1/2, which weakened osimertinib sensitivity in NSCLC cells. Consequently, this study aimed to identify a novel inhibitor which could suppress resistant cell growth by dual targeting of MEK1 and AKT1/2. Based on computational screening, we identified that costunolide could interact with MEK1 and AKT1/2. Further exploration using in vitro kinase assays validated that costunolide inhibited the kinase activity of MEK1 and AKT1/2, which restrained downstream ERK-RSK2 and GSK3ß signal transduction and significantly induced cell apoptosis. Remarkably, the combination of osimertinib and costunolide showed synergistic or additive inhibitory effects on tumor growth in osimertinib-resistant cell lines and PDX model. Hence, this study highlights a potential therapeutic strategy for osimertinib-resistant patients through targeting of MEK1 and AKT1/2 by costunolide.


Subject(s)
ErbB Receptors , Lung Neoplasms , Acrylamides , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Glycogen Synthase Kinase 3 beta/genetics , Humans , Indoles , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/genetics , Pyrimidines , Sesquiterpenes
2.
Nat Commun ; 13(1): 5651, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36163440

ABSTRACT

Knowing the mutation frequency of cancer genes in China is crucial for reducing the global health burden. We integrate the tumor epidemiological statistics with cancer gene mutation rates identified in 11,948 cancer patients to determine their weighted proportions within a Chinese cancer patient cohort. TP53 (51.4%), LRP1B (13.4%), PIK3CA (11.6%), KRAS (11.1%), EGFR (10.6%), and APC (10.5%) are identified as the top mutated cancer genes in China. Additionally, 18 common cancer types from both China and U.S. cohorts are analyzed and classified into three patterns principally based upon TP53 mutation rates: TP53-Top, TP53-Plus, and Non-TP53. Next, corresponding similarities and prominent differences are identified upon comparing the mutational profiles from both cohorts. Finally, the potential population-specific and environmental risk factors underlying the disparities in cancer gene mutation rates between the U.S. and China are analyzed. Here, we show and compare the mutation rates of cancer genes in Chinese and U.S. population cohorts, for a better understanding of the associated etiological and epidemiological factors, which are important for cancer prevention and therapy.


Subject(s)
Mutation Rate , Neoplasms , Class I Phosphatidylinositol 3-Kinases/genetics , ErbB Receptors/genetics , Genes, Neoplasm , Humans , Mutation , Neoplasms/epidemiology , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics
3.
J Cancer ; 13(8): 2607-2619, 2022.
Article in English | MEDLINE | ID: mdl-35711839

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a malignant cancer that is responsible for a high mortality rate; it accounts for approximately 90% of the 456,000 esophageal cancer (EC) cases diagnosed annually. Effective natural or synthesized compounds to prevent, treat, and/or inhibit ESCC relapse are desperately needed. The natural di-indole compound 3,3'-diindolylmethane (DIM) is abundant in cruciferous vegetables and shows potent anti-tumor effects in multiple cancers. The synthesized Eflornithine (DFMO) is clinically used to treat African sleeping sickness. We demonstrated that the combination of DIM+DFMO could significantly suppress the ESCC growth in the in vivo study of three patient-derived xenograft (PDX) cases. Then, the corresponding underlying anticancer mechanisms were investigated via the isobaric tags for relative and absolute quantification (iTRAQ) on the proteome level. We found that the DNA Replication and Cell Cycle were the top-2 most significantly downregulated signaling pathways following the DIM+DFMO treatment. Correspondingly and interestingly, these two pathways were the top-2 upregulated ones in clinic ESCC tumors. Moreover, the involved differentially expressed genes (DEGs) including MCM2, MCM3, MCM5, MCM6, MCM7, CDK1, and LIG1 were all inversely downregulated by DIM+DFMO treatment. In the limited clinical study in two ESCC cases, the administration of DIM (250mg) +DFMO (500 mg) once daily showed favorable results, including alleviated swallowing difficulties, decreased blood tumor markers (CA19-9, CA15-3 and AFP), and no severe toxicity in at least one month progression free survival period. We concluded that DIM+DFMO is a promising therapeutic combination for ESCC treatment via the suppression of DNA Replication and Cell Cycle activities. However, these therapeutic effects should be verified in large cohort clinical trials with sufficient cases.

4.
Cell Oncol (Dordr) ; 45(1): 179-191, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35102488

ABSTRACT

BACKGROUND: Hippocalcin-like 1 (HPCAL1), a neuronal calcium sensor protein family member, has been reported to regulate cancer growth. As yet, however, the biological functions of HPCAL1 and its molecular mechanisms have not been investigated in non-small cell lung carcinoma (NSCLC). METHODS: HPCAL1 expression in NSCLC samples was detected using immunohistochemistry, Western blotting and RT-PCR. The anticancer effects of HPCAL1 knockdown were determined by MTT, soft agar, cell cycle, oxygen consumption and reactive oxygen species assays. The effect of HPCAL1 knockdown on in vivo tumor growth was assessed using NSCLC cancer patient-derived xenograft models. Potentially interacting protein partners of HPCAL1 were identified using IP-MS/MS, immunoprecipitation and Western blotting assays. Metabolic alterations resulting from HPCAL1 knockdown were investigated using non-targeted metabolomics and RNA sequencing analyses. RESULTS: We found that HPCAL1 is highly expressed in NSCLC tissues and is positively correlated with low survival rates and AJCC clinical staging in lung cancer patients. Knockdown of HPCAL1 strongly increased oxygen consumption rates and the production of reactive oxygen species. HPCAL1 knockdown also inhibited NSCLC cell growth and patient-derived NSCLC tumor growth in vivo. Mechanistically, we found that HPCAL1 can directly bind to LDHA and enhance SRC-mediated phosphorylation of LDHA at tyrosine 10. The metabolomics and RNA sequencing analyses indicated that HPCAL1 knockdown reduces amino acid levels and induces fatty acid synthesis through regulating the expression of metabolism-related genes. Additionally, rescued cells expressing wild-type or mutant LDHA in HPCAL1 knockdown cells suggest that LDHA may serve as the main substrate of HPCAL1. CONCLUSIONS: Our data indicate that the effect of HPCAL1 knockdown on reducing SRC-mediated LDHA activity attenuates NSCLC growth. Our findings reveal novel biological functions and a mechanism underlying the role of HPCAL1 in NSCLC growth in vitro and in vivo.


Subject(s)
Carcinoma, Non-Small-Cell Lung , L-Lactate Dehydrogenase/metabolism , Lung Neoplasms , Neurocalcin/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Hippocalcin/genetics , Hippocalcin/metabolism , Humans , Lung Neoplasms/pathology , Tandem Mass Spectrometry
5.
Clin Transl Med ; 11(10): e548, 2021 10.
Article in English | MEDLINE | ID: mdl-34709754

ABSTRACT

RATIONALE: A high risk of post-operative recurrence contributes to the poor prognosis and low survival rate of oesophageal squamous cell carcinoma (ESCC) patients. Increasing experimental evidence suggests that integrin adhesion receptors, in particular integrin αv (ITGAV), are important for cancer cell survival, proliferation and migration. Therefore, targeting ITGAV may be a rational approach for preventing ESCC recurrence. MATERIALS AND METHODS: Protein levels of ITGAV were determined in human ESCC tumour tissues using immunohistochemistry. MTT, propidium iodide staining, and annexin V staining were utilized to investigate cell viability, cell cycle progression, and induction of apoptosis, respectively. Computational docking was performed with the Schrödinger Suite software to visualize the interaction between indomethacin and ITGAV. Cell-derived xenograft mouse models, patient-derived xenograft (PDX) mouse models, and a humanized mouse model were employed for in vivo studies. RESULTS: ITGAV was upregulated in human ESCC tumour tissues and increased ITGAV protein levels were associated with poor prognosis. ITGAV silencing or knockout suppressed ESCC cell growth and metastatic potential. Interestingly, we identified that indomethacin can bind to ITGAV and enhance synovial apoptosis inhibitor 1 (SYVN1)-mediated degradation of ITGAV. Integrin ß3, one of the ß subunits of ITGAV, was also decreased at the protein level in the indomethacin treatment group. Importantly, indomethacin treatment suppressed ESCC tumour growth and prevented recurrence in a PDX mouse model. Moreover, indomethacin inhibited the activation of cytokine TGFß, reduced SMAD2/3 phosphorylation, and increased anti-tumour immune responses in a humanized mouse model. CONCLUSION: ITGAV is a promising therapeutic target for ESCC. Indomethacin can attenuate ESCC growth through binding to ITGAV, promoting SYVN1-mediated ubiquitination of ITGAV, and potentiating cytotoxic CD8+ T cell responses.


Subject(s)
Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Indomethacin/pharmacology , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/metabolism , Neoplasm Recurrence, Local/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Heterografts , Humans , Mice, Nude
6.
J Cancer ; 12(13): 3930-3944, 2021.
Article in English | MEDLINE | ID: mdl-34093800

ABSTRACT

Esophageal Squamous Cell Carcinoma (ESCC) is the predominant type of Esophageal Cancer (EC), accounting for nearly 88% of EC incidents worldwide. Importantly, it is also a life-threatening cancer for patients diagnosed in advanced stages, with only a 20% 5-year survival rate due to a limited number of actionable targets and therapeutic options. Increasing evidence has shown that inter-tumor and intra-tumor heterogeneity are widely distributed across ESCC tumor tissues. In our work, multi-omics data from ESCC cell lines, tumor tissue, normal tissue and Patient-Derived Xenograft (PDX) tissues were analyzed to investigate the heterogeneity among ESCC samples at the DNA, RNA, and protein level. We identified enrichment of ECM-receptor interaction and Focal adhesion pathways from the subset of protein-coding genes with non-silent mutations in ESCC patients. We also found that TP53, TTN, KMT2D, CSMD3, DNAH5, MUC16 and DST are the most frequently mutated genes in ESCC patient samples. Out of the identified genes, TP53 is the most frequently mutated, with 84 distinct non-silent mutation variants. We observed that p.R248Q, p.R175G/H, and p.R273C/H are the most common TP53 mutation variants. The diversity of TP53 mutations reveal its importance in ESCC progression and may also provide promising targets for precision therapeutics. Additionally, we identified the Olfactory transduction as the top signaling pathway, enriched from genes uniquely expressed in The Cancer Genome Atlas (TCGA)-ESCC patient tumor tissues, which may provide implications for the exact roles of the corresponding genes in ESCC. Cyclic nucleotide-gated channel subunit beta 1(CNGB1), a gene belonging to the Olfactory transduction pathway, was found exclusively overexpressed in ESCC. Expression of CNGB1 could serve as a marker, indicating potential diagnostic or therapeutic value. Finally, we investigated heterogeneity in the context of the ESCC PDX model, which is an emerging tool used to predict drug response and recapitulate tumor behavior in vivo. We observed trans-species heterogeneity in as high as 75% of the identified proteins, indicating that the ambiguity of proteins should be addressed by specific strategies to avoid drawing false conclusions. The identification and characterization of gene mutation and expression heterogeneity across different ESCC datasets, including various novel TP53 mutations, ECM-receptor interaction, Focal adhesion, and Olfactory transduction pathways (CNGB1), provide researchers with evidence and implications for accurate research and precision therapeutic development.

7.
Crit Rev Immunol ; 40(6): 513-526, 2020.
Article in English | MEDLINE | ID: mdl-33900695

ABSTRACT

Currently the epidemic of SARS-CoV-2-caused COVID-19 is a major threat to global public health. The latest clinical data, laboratory results, and autopsy information are summarized herein to provide a brief review of the significant issues surrounding SARS-CoV-2 and COVID-19. In this review, we also cover research on the ways in which the virus enters the human body, general clinical symptoms, immunopathological responses in severe cases of COVID-19, and the issues surrounding the potential therapeutic responses to the illness.


Subject(s)
COVID-19/therapy , COVID-19/virology , SARS-CoV-2/physiology , COVID-19/pathology , Disease Management , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans
8.
Front Immunol ; 11: 621441, 2020.
Article in English | MEDLINE | ID: mdl-33584719

ABSTRACT

Although COVID-19 has become a major challenge to global health, there are currently no efficacious agents for effective treatment. Cytokine storm syndrome (CSS) can lead to acute respiratory distress syndrome (ARDS), which contributes to most COVID-19 mortalities. Research points to interleukin 6 (IL-6) as a crucial signature of the cytokine storm, and the clinical use of the IL-6 inhibitor tocilizumab shows potential for treatment of COVID-19 patient. In this study, we challenged wild-type and adenovirus-5/human angiotensin-converting enzyme 2-expressing BALB/c mice with a combination of polyinosinic-polycytidylic acid and recombinant SARS-CoV-2 spike-extracellular domain protein. High levels of TNF-α and nearly 100 times increased IL-6 were detected at 6 h, but disappeared by 24 h in bronchoalveolar lavage fluid (BALF) following immunostimulant challenge. Lung injury observed by histopathologic changes and magnetic resonance imaging at 24 h indicated that increased TNF-α and IL-6 may initiate CSS in the lung, resulting in the continual production of inflammatory cytokines. We hypothesize that TNF-α and IL-6 may contribute to the occurrence of CSS in COVID-19. We also investigated multiple monoclonal antibodies (mAbs) and inhibitors for neutralizing the pro-inflammatory phenotype of COVID-19: mAbs against IL-1α, IL-6, TNF-α, and granulocyte-macrophage colony-stimulating factor (GM-CSF), and inhibitors of p38 and JAK partially relieved CSS; mAbs against IL-6, TNF-α, and GM-CSF, and inhibitors of p38, extracellular signal-regulated kinase, and myeloperoxidase somewhat reduced neutrophilic alveolitis in the lung. This novel murine model opens a biologically safe, time-saving avenue for clarifying the mechanism of CSS/ARDS in COVID-19 and developing new therapeutic drugs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , COVID-19/immunology , Cytokine Release Syndrome/immunology , Disease Models, Animal , Spike Glycoprotein, Coronavirus/immunology , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , Cytokine Release Syndrome/virology , Cytokines/immunology , Male , Mice , Mice, Inbred BALB C , Poly I-C/immunology , SARS-CoV-2/immunology
9.
BMJ Open ; 9(6): e029160, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31230030

ABSTRACT

INTRODUCTION: Stroke is a leading cause of disability worldwide. The average hospital length of stay ranges from 3 to 28 days, and after discharge home the stroke survivors will live with physical, cognitive, even psychological disorders for the rest of their lives. It is essential to review the unmet needs of stroke survivors. METHODS AND ANALYSIS: A systematic review of previous quantitative and qualitative studies reporting the unmet needs of stroke survivors in their homes will be conducted. The following six databases will be searched from inception to December 2018 for relevant articles: PubMed, EMBASE, CINAHL, PsycINFO, SCOPUS and China Biology Medicine. We will include studies limited to human and published in English or Chinese, and the patients with stroke should discharge home rather than any other professional organisations including nursing homes or community rehabilitation units and so on. Data of quantitative research will be standardised for comparison, thematic analysis will be used for qualitative data and a narrative synthesis and pooled analysis of the main outcomes will be reported. ETHICS AND DISSEMINATION: This review will be submitted to an international professional journal, and the detailed search strategies and analysis flowchart will be openly included as supplements. This study does not require ethical approval as no patient's identifiable data will be used. Our findings will give a new look at the aspect of stroke survivors' unmet needs in their long-term recovery stage, especially the trajectories of unmet needs at different timepoints. What is more, this review will demonstrate the long-term unmet needs of stroke survivors from different countries, will compare any variations between high-income and low-income regions, and the geographical differences of needs will be mapped if necessary. We will endeavour to provide as much information as possible to healthcare professionals and public health policy makers in order to promote further medical reform. TRIAL REGISTRATION NUMBER: CRD42018112181.


Subject(s)
Independent Living , Needs Assessment , Stroke Rehabilitation , Survivors/psychology , Activities of Daily Living , Adaptation, Psychological , Cognition , Humans , Independent Living/psychology , Independent Living/standards , Research Design , Stroke Rehabilitation/methods , Stroke Rehabilitation/psychology , Systematic Reviews as Topic
10.
Cancer Lett ; 448: 20-30, 2019 04 28.
Article in English | MEDLINE | ID: mdl-30716361

ABSTRACT

3,3'-Diindolymethane (DIM) is a dimeric condensation product of indole-3-carbinol (I3C) that is found in broccoli and cabbage. Although DIM has been reported to exhibit anticancer properties against multiple tumor types, the direct target proteins of DIM have not been fully investigated. In the present study, we report that DIM is a novel COX1/2 and ERK1/2 inhibitor that suppresses growth of colon cancer in vitro and in vivo. To identify possible molecular targets of DIM, 11 potential candidate proteins were validated by an in vitro kinase or enzyme assay. We found that DIM directly inhibits COX1/2 and ERK1/2 protein activities in vitro. Additionally, the PGE2 production (COX-mediated metabolite) and phosphorylated RSK expression (ERK1/2 direct downstream kinase) were strongly suppressed by DIM in colon cancer cells. The inhibition of cell growth by DIM is dependent on the expression of COX1/2 or ERK1/2 proteins. Notably, oral administration of DIM suppressed patient-derived xenograft colon tumor growth in an in vivo mouse model. Overall these results suggest that DIM is a potent and dual COX1/2 and ERK1/2 inhibitor that might be used for chemotherapy against colon cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Cyclooxygenase 2/metabolism , Humans , Mice , Signal Transduction/drug effects , Tumor Cells, Cultured
11.
Phytother Res ; 33(3): 640-650, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30536456

ABSTRACT

Gossypin is a flavone extracted from Hibiscus vitifolius, which has been reported to exhibit anti-inflammatory, antioxidant, and anticancer activities. However, the anticancer properties of gossypin and its molecular mechanism of action against gastric cancer have not been fully investigated. In the present study, we report that gossypin is an Aurora kinase A (AURKA) and RSK2 inhibitor that suppresses gastric cancer growth. Gossypin attenuated anchorage-dependent and anchorage-independent gastric cancer cell growth as well as cell migration. Based on the results of in vitro screening and cell-based assays, gossypin directly binds to and inhibits AURKA and RSK2 activities and their downstream signaling proteins. Gossypin decreased S phase and increased G2/M phase cell cycle arrest by reducing the expression of cyclin A2 and cyclin B1 and the phosphorylation of the CDC protein. Additionally, gossypin also induced intrinsic apoptosis by activating caspases and PARP and increasing the expression of cytochrome c. Our results demonstrate that gossypin is an AURKA and RSK2 inhibitor that could be useful for treating gastric cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Flavonoids/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Humans , Stomach Neoplasms/pathology
12.
Int J Nurs Pract ; 24(2): e12625, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29457315

ABSTRACT

AIMS: To develop a culturally appropriate and functional standard Mandarin Chinese translation of the Self-Administration of Medication tool and to examine its validity and reliability. METHODS: We used Brislin's guidelines for the translation and back-translation procedures. We recruited 130 patients from June 2015 to September 2016. Correlation analysis, Cronbach's alpha coefficient, split-half reliability, item analysis and the content validity index, and exploratory factor analysis were performed. RESULTS: Strong and moderate correlations were seen between the total Self-Administration of Medication score and nurses' and patients' perceptions. Item analysis indicated that the correlation coefficient ranged from 0.744 to 0.975. Cronbach's alpha ranged from 0.933 to 0.976. The split-half reliability ranged from 0.911 to 0.936 for the instrument's subscales, and 0.953 for the total scale. The content validity index value ranged from 0.823 to 0.972 for individual items and was 0.968 for the total scale. The factor loading matrix of the SAM ranged from 0.514 to 0.837. A moderate correlation existed between the scores of the Self-Administration of Medication tool and the Barthel Index. CONCLUSION: The Chinese version of the Self-Administration of Medication tool is statistically acceptable and can be used in the evaluation of self-medication ability based on self-report and nurses' assessment.


Subject(s)
Chronic Disease/drug therapy , Self Administration , Surveys and Questionnaires , Aged , China , Female , Humans , Male , Middle Aged , Psychometrics , Reproducibility of Results , Translations
13.
Carcinogenesis ; 38(11): 1136-1146, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29029040

ABSTRACT

Herbacetin is a flavonol compound that is found in plants such as flaxseed and ramose scouring rush herb, it possesses a strong antioxidant capacity, and exerts anticancer effects on colon and breast cancer. However, the effect of herbacetin on skin cancer has not been investigated. Herein, we identified herbacetin as a dual V-akt murine thymoma viral oncogene homolog (AKT) and ornithine decarboxylase (ODC) inhibitor, and illustrated its anticancer effects in vitro and in vivo against cutaneous squamous cell carcinoma (SCC) and melanoma cell growth. To identify the direct target(s) of herbacetin, we screened several skin cancer-related protein kinases, and results indicated that herbacetin strongly suppresses both AKT and ODC activity. Results of cell-based assays showed that herbacetin binds to both AKT and ODC, inhibits TPA-induced neoplastic transformation of JB6 mouse epidermal cells, and suppresses anchorage-independent growth of cutaneous SCC and melanoma cells. The inhibitory activity of herbacetin was associated with markedly reduced NF-κB and AP1 reporter activity. Interestingly, herbacetin effectively attenuated TPA-induced skin cancer development and also exhibited therapeutic effects against solar-UV-induced skin cancer and melanoma growth in vivo. Our findings indicate that herbacetin is a potent AKT and ODC inhibitor that should be useful for preventing skin cancers.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Flavonoids/pharmacology , Melanoma/drug therapy , Ornithine Decarboxylase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/drug therapy , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Humans , Melanoma/metabolism , Mice , NF-kappa B , Ornithine Decarboxylase Inhibitors/pharmacology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL