1
|
Akhter N, Dar SA, Chattopadhyay S, Haque S, Anwer R, Wahid M, Jawed A, Lohani M, Mandal RK, Shukla NK, Abdul Y, Husain SA. Impact of p53 arg72pro SNP on Breast Cancer Risk in North Indian Population. Curr Genomics 2018; 19:395-410. [PMID: 30065615 PMCID: PMC6030857 DOI: 10.2174/1389202919666171205104137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 12/31/2022] Open
Abstract
Background: Genetic changes in p53 gene contribute to breast cancer susceptibility. Objective and Methods: A case-control study and a meta-analysis were performed to investigate the role of p53 codon72 SNP with breast cancer susceptibility in Indian women. Results: p53 heterozygous arginine variant was associated with decreased risk of breast cancer in total cohort. In meta-analysis, Allelic and GG vs. CC genetic comparison model were found to be associated with breast cancer risk. Moreover, recessive comparison model indicated a protective correlation with breast cancer occurrence. Conclusion: The findings of our case-control study and meta-analysis suggest a significant association between p53 Arg72Pro polymorphism and an increased risk of breast cancer in Indian population.
Collapse
Affiliation(s)
- Naseem Akhter
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia.,Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| | - Sajad A Dar
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia
| | - Shilpi Chattopadhyay
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| | - Shafiul Haque
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India.,Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia
| | - Razique Anwer
- Department of Anatomy, College of Medicine, Al Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Mohd Wahid
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India.,Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia
| | - Arshad Jawed
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia
| | - Mohtashim Lohani
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia.,Department of Biosciences, Integral University, Lucknow - 226026, Uttar Pradesh, India
| | - Raju K Mandal
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia
| | | | | | - Syed Akhtar Husain
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| |
Collapse
|
2
|
Samuelsson LB, Bovbjerg DH, Roecklein KA, Hall MH. Sleep and circadian disruption and incident breast cancer risk: An evidence-based and theoretical review. Neurosci Biobehav Rev 2017; 84:35-48. [PMID: 29032088 DOI: 10.1016/j.neubiorev.2017.10.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 09/26/2017] [Accepted: 10/11/2017] [Indexed: 02/08/2023]
Abstract
Opportunities for restorative sleep and optimal sleep-wake schedules are becoming luxuries in industrialized cultures, yet accumulating research has revealed multiple adverse health effects of disruptions in sleep and circadian rhythms, including increased risk of breast cancer. The literature on breast cancer risk has focused largely on adverse effects of night shift work and exposure to light at night (LAN), without considering potential effects of associated sleep disruptions. As it stands, studies on breast cancer risk have not considered the impact of both sleep and circadian disruption, and the possible interaction of the two through bidirectional pathways, on breast cancer risk in the population at large. We review and synthesize this literature, including: 1) studies of circadian disruption and incident breast cancer; 2) evidence for bidirectional interactions between sleep and circadian systems; 3) studies of sleep and incident breast cancer; and 4) potential mechanistic pathways by which interrelated sleep and circadian disruption may contribute to the etiology of breast cancer.
Collapse
Affiliation(s)
- Laura B Samuelsson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dana H Bovbjerg
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Department of Behavioral & Community Health Sciences, University of Pittsburgh, Pittsburgh, PA, United States; Biobehavioral Oncology Program, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kathryn A Roecklein
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Martica H Hall
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
3
|
Israr M, Biryukov J, Ryndock EJ, Alam S, Meyers C. Comparison of human papillomavirus type 16 replication in tonsil and foreskin epithelia. Virology 2016; 499:82-90. [PMID: 27639574 DOI: 10.1016/j.virol.2016.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/29/2016] [Accepted: 09/06/2016] [Indexed: 12/24/2022]
Abstract
Human papillomavirus (HPV) is well recognized as a causative agent for anogenital and oropharyngeal cancers, however, the biology of HPV infection at different mucosal locations, specifically the oral cavity, is not well understood. Importantly, it has yet to be determined if oral tissues are permissive for HPV infection and replication. We investigated for the first time the titers, infectivity, and maturation of HPV16 in oral epithelial versus genital epithelial tissue. We show that infectious HPV16 virions can be produced in oral tissue. This demonstrates, for the first time, that infectious virus could be spread via the oral cavity. HPV16 derived from oral tissue utilize a tissue-spanning redox gradient that facilitates the maturation of virions over time. Maturation is manifested by virion stability and increased susceptibility to neutralization with anti-HPV16 L1 antibodies. However, susceptibility to neutralization by anti-HPV16 L2 specific antibodies decreases during the maturation of HPV16 virions in oral tissue.
Collapse
Affiliation(s)
- Mohd Israr
- Department of Pediatrics, Division of Allergy and Immunology, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Jennifer Biryukov
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Eric J Ryndock
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Samina Alam
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Craig Meyers
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
4
|
Rowson-Hodel AR, Manjarin R, Trott JF, Cardiff RD, Borowsky AD, Hovey RC. Neoplastic transformation of porcine mammary epithelial cells in vitro and tumor formation in vivo. BMC Cancer 2015; 15:562. [PMID: 26228788 PMCID: PMC4520266 DOI: 10.1186/s12885-015-1572-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 07/17/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mammary glands of pigs share many functional and morphological similarities with the breasts of humans, raising the potential of their utility for research into the mechanisms underlying normal mammary function and breast carcinogenesis. Here we sought to establish a model for the efficient manipulation and transformation of porcine mammary epithelial cells (pMEC) in vitro and tumor growth in vivo. METHODS We utilized a vector encoding the red florescent protein tdTomato to transduce populations of pMEC from Yorkshire -Hampshire crossbred female pigs in vitro and in vivo. Populations of primary pMEC were then separated by FACS using markers to distinguish epithelial cells (CD140a-) from stromal cells (CD140a+), with or without further enrichment for basal and luminal progenitor cells (CD49f+). These separated pMEC populations were transduced by lentivirus encoding murine polyomavirus T antigens (Tag) and tdTomato and engrafted to orthotopic or ectopic sites in immunodeficient NOD.Cg-Prkdc (scid) Il2rg (tm1Wjl) /SzJ (NSG) mice. RESULTS We demonstrated that lentivirus effectively transduces pMEC in vitro and in vivo. We further established that lentivirus can be used for oncogenic-transformation of pMEC ex vivo for generating mammary tumors in vivo. Oncogenic transformation was confirmed in vitro by anchorage-independent growth, increased cell proliferation, and expression of CDKN2A, cyclin A2 and p53 alongside decreased phosphorylation of Rb. Moreover, Tag-transformed CD140a- and CD140a-CD49f + pMECs developed site-specific tumors of differing histopathologies in vivo. CONCLUSIONS Herein we establish a model for the transduction and oncogenic transformation of pMEC. This is the first report describing a porcine model of mammary epithelial cell tumorigenesis that can be applied to the study of human breast cancers.
Collapse
Affiliation(s)
- A R Rowson-Hodel
- Department of Animal Science, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Present Address: Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, USA.
| | - R Manjarin
- Department of Animal Science, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Present Address: USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA.
| | - J F Trott
- Department of Animal Science, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - R D Cardiff
- Center for Comparative Medicine, University of California Davis, One Shields Avenue, Davis, CA, USA.
| | - A D Borowsky
- Center for Comparative Medicine, University of California Davis, One Shields Avenue, Davis, CA, USA.
| | - R C Hovey
- Department of Animal Science, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
5
|
Kitazono M, Shinchi H, Ishigami S, Ueno S, Natsugoe S. Effects of a histone deacetylase inhibitor, sodium butyrate, on 53-kDa protein expression and sensitivity to anticancer drugs of pancreatic cancer cells. Curr Ther Res Clin Exp 2014; 71:162-72. [PMID: 24683262 DOI: 10.1016/j.curtheres.2010.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2010] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Several tumor-suppressor genes, such as 53-kDa protein (p53), are inactivated in some pancreatic cancers. The lack of a functional p53 has been proposed to be a component of resistance to chemotherapy, resulting in the inhibition of apoptosis. Therefore, reintroduction of wild-type p53 is a commonly used gene therapy strategy for the treatment of various types of cancer, including pancreatic cancer. OBJECTIVE The aim of this study was to examine the ability of the histone deacetylase inhibitor, sodium butyrate (NaB), to modulate the expression of p53. METHODS Five human pancreatic carcinoma cell lines (SW-1990, BxPC-3, PANC-1, MIA PaCa-2, JHP-1) were utilized. Two of the cell lines (SW-1990 and JHP-1) lacked p53 expression, as determined by Western blot analysis, and were investigated further. Expression of p53 was determined by densitometry of all bands present in the Western blot. Drug sensitivity was measured with a tetrazolium-based assay by exposing the cells to graded concentrations of NaB and/or anticancer drugs (cisplatin, fluorouracil, SN-38, and paclitaxel). Apoptosis was observed using gel electrophoresis. RESULTS In the SW-1990 and JHP-1 cell lines, use of 1 mM NaB was found to induce histone acetylation and p53 expression compared with those not treated with NaB (P = 0.01 and P = 0.018, respectively). Sensitivity to cisplatin (P = 0.021), fluorouracil (P = 0.046), and SN-38 (P = 0.039) was significantly enhanced by NaB treatment compared with nontreatment. However, sensitivity to paclitaxel was not significantly different between untreated and NaB-treated cells. A higher frequency of apoptosis was observed in NaB-treated cells compared with that of control cells. CONCLUSION This in vitro study found that NaB induced p53 expression in 2 pancreatic cancer cell lines (SW-1990 and JHP-1). Moreover, NaB acted on a biochemical modulator for antieuplastic therapy. Future research is necessary to assess the value of these findings.
Collapse
Affiliation(s)
- Masaki Kitazono
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Hiroyuki Shinchi
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Sumiya Ishigami
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Shinichi Ueno
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Shoji Natsugoe
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| |
Collapse
|
6
|
Newton HB. Molecular neuro-oncology and the development of targeted therapeutic strategies for brain tumors Part 4: p53 signaling pathway. Expert Rev Anticancer Ther 2014; 5:177-91. [PMID: 15757449 DOI: 10.1586/14737140.5.1.177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain tumors are a diverse group of malignancies that remain refractory to conventional treatment approaches. Molecular neuro-oncology has now begun to clarify the transformed phenotype of brain tumors and identify oncogenic pathways that might be amenable to targeted therapy. Loss of the tumor suppressor gene p53 and its encoded protein are the most common genetic events in human cancer and are a frequent occurrence in brain tumors. p53 functions as a transcription factor and is responsible for the transactivation and repression of key genes involved in cell growth, apoptosis and the cell cycle. Mutation of the p53 gene or dysfunction of its signaling pathway are early events in the transformation process of astrocytic gliomas. The majority of mutations are missense and occur in the conserved regions of the gene, within exons 5 through 8. Molecular therapeutic strategies to normalize p53 signaling in cells with mutant p53 include pharmacologic rescue of mutant protein, gene therapy approaches, small-molecule agonists of downstream inhibitory genes, antisense approaches and oncolytic viruses. Other strategies include activation of normal p53 activity, inhibition of mdm2-mediated degradation of p53 and blockade of p53 nuclear export. Further development of targeted therapies designed to restore or enhance p53 function, and evaluation of these new agents in clinical trials, will be needed to improve survival and quality of life for patients with brain tumors.
Collapse
Affiliation(s)
- Herbert B Newton
- Dardinger Neuro-Oncology Center, Department of Neurology, Ohio State University Hospitals, 465 Means Hall, 1654 Upham Drive, Columbus, OH 43210, USA.
| |
Collapse
|
7
|
PRASAD GANAPATHYA, WANG KENNETHK, HALLING KEVINC, BUTTAR NAVTEJS, WONGKEESONG LOUIS, ZINSMEISTER ALANR, BRANKLEY SHANNONM, BARR FRITCHER EMILYG, WESTRA WYTSKEM, KRISHNADATH KAUSILIAK, LUTZKE LORIS, BORKENHAGEN LYNNS. Utility of biomarkers in prediction of response to ablative therapy in Barrett's esophagus. Gastroenterology 2008; 135:370-9. [PMID: 18538141 PMCID: PMC3896328 DOI: 10.1053/j.gastro.2008.04.036] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 03/31/2008] [Accepted: 04/30/2008] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Photodynamic therapy (PDT) has been shown to be effective in the treatment of high-grade dysplasia (HGD)/mucosal carcinoma in Barrett's esophagus (BE). Substantial proportions of patients do not respond to PDT or progress to carcinoma despite PDT. The role of biomarkers in predicting response to PDT is unknown. We aimed to determine if biomarkers known to be associated with neoplasia in BE can predict loss of dysplasia in patients treated with ablative therapy for HGD/intramucosal cancer. METHODS Patients with BE and HGD/intramucosal cancer were studied prospectively from 2002 to 2006. Biomarkers were assessed using fluorescence in situ hybridization performed on cytology specimens, for region-specific and centromeric probes. Patients were treated with PDT using cylindric diffusing fibers (wavelength, 630 nm; energy, 200 J/cm fiber). Univariate and multiple variable logistic regression was performed to determine predictors of response to PDT. RESULTS A total of 126 consecutive patients (71 who underwent PDT and 55 patients who did not undergo PDT and were under surveillance, to adjust for the natural history of HGD), were included in this study. Fifty (40%) patients were responders (no dysplasia or carcinoma) at 3 months after PDT. On multiple variable analysis, P16 allelic loss (odds ratio [OR], 0.32; 95% confidence interval [CI], 0.10-0.96) predicted decreased response to PDT. BE segment length (OR, 0.71; 95% CI, 0.59-0.85), and performance of PDT (OR, 7.17; 95% CI, 2.50-20.53) were other independent predictors of loss of dysplasia. CONCLUSIONS p16 loss detected by fluorescence in situ hybridization can help predict loss of dysplasia in patients with BE and HGD/mucosal cancer. Biomarkers may help in the selection of appropriate therapy for patients and improve treatment outcomes.
Collapse
Affiliation(s)
- GANAPATHY A. PRASAD
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - KENNETH K. WANG
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - KEVIN C. HALLING
- Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - NAVTEJ S. BUTTAR
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - LOUIS–MICHEL WONGKEESONG
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - ALAN R. ZINSMEISTER
- Division of Biostatistics, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - SHANNON M. BRANKLEY
- Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - EMILY G. BARR FRITCHER
- Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - WYTSKE M. WESTRA
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - KAUSILIA K. KRISHNADATH
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - LORI S. LUTZKE
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - LYNN S. BORKENHAGEN
- Barrett’s Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| |
Collapse
|
8
|
Chhipa RR, Kumari R, Upadhyay AK, Bhat MK. Abrogation of p53 by its antisense in MCF-7 breast carcinoma cells increases cyclin D1 via activation of Akt and promotion of cell proliferation. Exp Cell Res 2007; 313:3945-58. [PMID: 17935714 DOI: 10.1016/j.yexcr.2007.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 07/26/2007] [Accepted: 08/20/2007] [Indexed: 11/17/2022]
Abstract
The p53 protein has been a subject of intense research interest since its discovery as about 50% of human cancers carry p53 mutations. Mutations in the p53 gene are the most frequent genetic lesions in breast cancers suggesting a critical role of p53 in breast cancer development, growth and chemosensitivity. This report describes the derivation and characterization of MCF-7As53, an isogenic cell line derived from MCF-7 breast carcinoma cells in which p53 was abrogated by antisense p53 cDNA. Similar to MCF-7 and simultaneously selected hygromycin resistant MCF-7H cells, MCF-7As53 cells have consistent basal epithelial phenotype, morphology, and estrogen receptor expression levels at normal growth conditions. Present work documents investigation of molecular variations, growth kinetics, and cell cycle related studies in relation to absence of wild-type p53 protein and its transactivation potential as well. Even though wild-type tumor suppressor p53 is an activator of cell growth arrest and apoptosis-mediator genes such as p21, Bax, and GADD45 in MCF-7As53 cells, no alterations in expression levels of these genes were detected. The doubling time of these cells decreased due to depletion of G0/G1 cell phase because of constitutive activation of Akt and increase in cyclin D1 protein levels. This proliferative property was abrogated by wortmannin, an inhibitor of PI3-K/Akt signaling pathway. Therefore this p53 null cell line indicates that p53 is an indispensable component of cellular signaling system which is regulated by caveolin-1 expression, involving Akt activation and increase in cyclin D1, thereby promoting proliferation of breast cancer cells.
Collapse
Affiliation(s)
- Rishi Raj Chhipa
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, Pune-411 007, India
| | | | | | | |
Collapse
|
9
|
Zhang L, Levi E, Majumder P, Yu Y, Aboukameel A, Du J, Xu H, Mohammad R, Hatfield JS, Wali A, Adsay V, Majumdar APN, Rishi AK. Transactivator of transcription-tagged cell cycle and apoptosis regulatory protein-1 peptides suppress the growth of human breast cancer cells in vitro and in vivo. Mol Cancer Ther 2007; 6:1661-72. [PMID: 17513614 DOI: 10.1158/1535-7163.mct-06-0653] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Deregulated signaling by the epidermal growth factor receptor family of proteins is encountered in human malignancies including breast cancer. Cell cycle and apoptosis-regulatory protein-1 (CARP-1), a novel, perinuclear phosphoprotein, is a regulator of apoptosis signaling by epidermal growth factor receptors. CARP-1 expression is diminished in human breast cancers, and correlates inversely with human breast cancer grades which could be attributed to increased methylation. The expression of CARP-1, on the other hand, interferes with the ability of human breast cancer cells to invade through the matrigel-coated membranes, to form colonies in the soft agar, and to grow as s.c. tumors in severe combined immunodeficiency (SCID) mice. To test whether CARP-1 is a suppressor of human breast cancer growth, we generated transactivator of transcription (TAT)-tagged CARP-1 peptides. Treatment of human breast cancer cells with affinity purified, TAT-CARP-1 1-198, 197-454, and 896-1150 peptides caused inhibition of human breast cancer cell proliferation and elevated apoptosis. In contrast, TAT-tagged enhanced green fluorescent protein or CARP-1 (1-198(Y192/F)) peptide failed to inhibit cell proliferation or induce apoptosis. Apoptosis by CARP-1 peptides, with the exception of CARP-1 (1-198(Y192/F)), involves the activation of p38 stress-activated protein kinase and caspase-9. Moreover, administration of TAT-CARP-1 (1-198), but not TAT-tagged enhanced green fluorescent protein or TAT-CARP-1 (1-198(Y192/F)), inhibits growth of human breast cancer cell-derived tumor xenografts in SCID mice. We conclude that CARP-1 is a suppressor of human breast cancer growth, and its expression is diminished in tumors, in part, by methylation-dependent silencing.
Collapse
Affiliation(s)
- Liyue Zhang
- Veterans Affairs Medical Center, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Chhipa RR, Bhat MK. Bystander killing of breast cancer MCF-7 cells by MDA-MB-231 cells exposed to 5-fluorouracil is mediated via Fas. J Cell Biochem 2007; 101:68-79. [PMID: 17340621 DOI: 10.1002/jcb.21153] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The major drawback with cancer therapy is the development of resistant cells within tumors due to their heterogeneous nature and due to inadequate drug delivery during chemotherapy. Therefore, the propagation of injury ("bystander effect" (BE)) from directly damaged cells to other cells may have great implications in cancer chemotherapy. The general advantage of the bystander cell killing phenomenon is the large therapeutic index that can be achieved. Experiments suggest that this phenomenon is detected in radiation therapy as well as in gene therapy in conjunction with chemotherapy. In the present study, we developed an original in vitro model dedicated to the exploration of bystander cytotoxicity induced during breast carcinoma chemotherapy. In brief, we investigated this perpetuation of injury on untreated bystander MCF-7 breast cancer cells which were coplated with 5-fluorouracil (5-FU)-treated MDA-MB-231 breast cancer cells. To achieve this goal, a specific in vitro coculture model which involved mixing of aggressive MDA-MB-231 breast cancer cells with enhanced green fluorescent protein (EGFP) expressing stable clone of non-metastatic MCF-7 breast cancer cells (MCF-EGFP), was used. A bystander killing effect was observed in MCF-EGFP cells cocultured with MDA-MB-231 cells pretreated with 5-FU. The striking decrease in MCF-EGFP cells, as detected by assaying for total GFP intensity, is mediated by activation of Fas/FasL system. The implication of Fas in MCF-EGFP cell death was confirmed by using antagonistic anti-FasL antibody that reverses bystander cell death by blocking FasL on MDA-MB-231 cells. In addition, inhibition of CD95/Fas receptor on the cell surface of MCF-EGFP cells by treatment with Pifithrin-alpha, a p53 specific transactivation inhibitor, partially abrogated the sensitivity of bystander MCF-EGFP cells. Our data, therefore, demonstrates that the Fas/FasL system could be considered as a new determinant for chemotherapy-induced bystander cell death in breast cancers.
Collapse
Affiliation(s)
- Rishi Raj Chhipa
- National Centre for Cell Science, Ganeshkhind, Pune 411 007, India
| | | |
Collapse
|
11
|
Thirukkumaran CM, Russell JA, Stewart DA, Morris DG. Viral purging of haematological autografts: should we sneeze on the graft? Bone Marrow Transplant 2007; 40:1-12. [PMID: 17450184 DOI: 10.1038/sj.bmt.1705668] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
High-dose cytotoxic chemotherapy followed by autologous haematopoietic stem cell transplantation (ASCT) is extensively used for the treatment of many haematopoietic, as well as several epithelial cancers. Disease relapse may be the result of tumour contamination within autograft as evidenced by gene marking studies. The multiple purging strategies that have been described to date have not proven effective in most ASCT settings. This review addresses the possibility of using oncolytic viruses as a novel purging strategy. DNA viruses such as genetically engineered adenoviral vectors have widely been used to deliver either a prodrug-activating enzyme or express wild-type p53 selectively in tumour cells in ex vivo purging protocols. In addition, conditionally replicating adenoviruses that selectively replicate in tumour cells and herpes simplex virus type 1 are other DNA viruses that have been tested as ex vivo purging agents under laboratory conditions. Vesicular stomatitis virus (VSV) and reovirus are naturally occurring RNA viruses that appear to hold promise as purging agents under ex vivo and in vivo settings. Preclinical data demonstrate reovirus's purging potential against breast, monocytic and myeloma cell lines as well as patient-derived tumours of diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, Waldenstrom macroglobulinemia and small lymphocytic lymphoma. In addition, VSV has shown effective killing of leukaemic cell lines and multiple myeloma patient specimens. Given the increasing interest in the utilization of viruses as purging agents, the following review provides a timely summary of the potential and the challenges of oncolytic viruses as purging modalities during ASCT.
Collapse
Affiliation(s)
- C M Thirukkumaran
- Department of Medicine, Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
12
|
Bay BH, Jin R, Huang J, Tan PH. Metallothionein as a prognostic biomarker in breast cancer. Exp Biol Med (Maywood) 2006; 231:1516-21. [PMID: 17018874 DOI: 10.1177/153537020623100910] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the most common cancer in women, with a general upward trend in incidence. Basic and clinical breast cancer research has continued at a rapid pace, in the endeavor to understand the biology of the disease so as to improve management of patients. Besides traditional pathological indicators, expression of molecular markers in breast cancer has also been comprehensively investigated. This paper will focus on the prognostic utility of metallothioneins (MTs), a family of low molecular weight metal binding proteins encoded by at least 10 functional MT genes that are associated with cell proliferation in breast cancer. Evidence that MT is a potential prognostic biomarker for breast cancer is supported by many reports in the literature. Expression of the MT protein has been detected by immunohistochemistry in a significant portion of invasive ductal breast cancers. MT expression has also been well studied in association with traditional clinico-pathological parameters of breast cancers. Generally, higher MT expression in breast cancers is predictive of worse patient outcomes. The relationship of MT isoforms to histological grade, estrogen receptor (ER) status, and prognosis will also be discussed.
Collapse
Affiliation(s)
- Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 4 Medical Drive, Blk MD 10, Singapore 117 597.
| | | | | | | |
Collapse
|
13
|
Newton HB. Molecular neuro-oncology and the development of targeted therapeutic strategies for brain tumors. Part 5: apoptosis and cell cycle. Expert Rev Anticancer Ther 2006; 5:355-78. [PMID: 15877531 DOI: 10.1586/14737140.5.2.355] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Brain tumors are a diverse group of malignancies that remain refractory to conventional treatment approaches. Molecular neuro-oncology has now begun to clarify the transformed phenotype of brain tumors and identify oncogenic pathways that might be amenable to targeted therapy. Abnormalities of the apoptotic and cell cycle signaling pathways are key molecular features of many brain tumors and are currently under evaluation for potential therapeutic intervention. The apoptosis pathway has numerous targets for molecular therapeutic development, including p53, Bax, Bcl-2, cFLIP, effector caspases, growth factor receptors, phosphatidylinositol-3-kinase, Akt and apoptosis inhibitors. Current molecular treatment approaches include antisense techniques, gene therapy and small-molecule modulators and inhibitors. Potential targets of the cell cycle pathway include the cyclins, cyclin-dependent kinases, p53, retinoblastoma, E2F and the cyclin-dependent kinase inhibitors. Developmental molecular therapeutics for this pathway include adenoviral and gene therapy, small-peptide cyclin-dependent kinase modulators, proteasomal inhibitors and small-molecule cyclin-dependent kinase inhibitors. Several of these recently identified agents have begun evaluation in clinical trials. Further development of targeted therapies designed to modulate apoptosis and the cell cycle, and evaluation of these new agents in clinical trials, will be needed to improve survival and quality of life for patients with brain tumors.
Collapse
Affiliation(s)
- Herbert B Newton
- Dardinger Neuro-Oncology Center, Department of Neurology, The Ohio State University Hospitals, 465 Means Hall, 1654 Upham Drive, Columbus, OH 43210, USA.
| |
Collapse
|
14
|
Scian MJ, Stagliano KER, Anderson MAE, Hassan S, Bowman M, Miles MF, Deb SP, Deb S. Tumor-derived p53 mutants induce NF-kappaB2 gene expression. Mol Cell Biol 2005; 25:10097-110. [PMID: 16260623 PMCID: PMC1280285 DOI: 10.1128/mcb.25.22.10097-10110.2005] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Overexpression of mutant p53 is a common theme in tumors, suggesting a selective pressure for p53 mutation in cancer development and progression. To determine how mutant p53 expression may lead to survival advantage in human cancer cells, we generated stable cell lines expressing p53 mutants p53-R175H, -R273H, and -D281G by use of p53-null human H1299 (lung carcinoma) cells. Compared to vector-transfected cells, H1299 cells expressing mutant p53 showed a survival advantage when treated with etoposide, a common chemotherapeutic agent; however, cells expressing the transactivation-deficient triple mutant p53-D281G (L22Q/W23S) had significantly lower resistance to etoposide. Gene expression profiling of cells expressing transcriptionally active mutant p53 proteins revealed the striking pattern that all three p53 mutants induced expression of approximately 100 genes involved in cell growth, survival, and adhesion. The gene NF-kappaB2 is a prominent member of this group, whose overexpression in H1299 cells also leads to chemoresistance. Treatment of H1299 cells expressing p53-R175H with small interfering RNA specific for NF-kappaB2 made these cells more sensitive to etoposide. We have also observed activation of the NF-kappaB2 pathway in mutant p53-expressing cells. Thus, one possible pathway through which mutants of p53 may induce loss of drug sensitivity is via the NF-kappaB2 pathway.
Collapse
Affiliation(s)
- Mariano J Scian
- Department of Biochemistry and Massey Cancer Center, Virginia Commonwealth University, P.O. Box 980614, Richmond, Virginia 23298, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Lin NH, Xia P, Kovar P, Park C, Chen Z, Zhang H, Rosenberg SH, Sham HL. Synthesis and biological evaluation of 3-ethylidene-1,3-dihydro-indol-2-ones as novel checkpoint 1 inhibitors. Bioorg Med Chem Lett 2005; 16:421-6. [PMID: 16242328 DOI: 10.1016/j.bmcl.2005.09.064] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 09/21/2005] [Accepted: 09/22/2005] [Indexed: 11/18/2022]
Abstract
Chk1 inhibitors have emerged as a novel class of neoplastic agents for abrogating the G2 DNA damage checkpoint arrest. Analogs of the Chk1 inhibitor, 3-ethylidene-1,3-dihydro-indol-2-one, were synthesized and tested in vitro for their inhibitory activities. The most promising compound identified from this series is analog 28, which possesses potent enzymatic and cellular activities.
Collapse
Affiliation(s)
- Nan-Horng Lin
- Cancer Research, R-47B, Global Pharmaceutical Products Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, IL 60064-3500, USA.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Bertucci F, Tarpin C, Charafe-Jauffret E, Bardou VJ, Braud AC, Tallet A, Gravis G, Viret F, Gonçalves A, Houvenaeghel G, Blaise D, Jacquemier J, Maraninchi D, Viens P. Multivariate analysis of survival in inflammatory breast cancer: impact of intensity of chemotherapy in multimodality treatment. Bone Marrow Transplant 2004; 33:913-20. [PMID: 15004544 DOI: 10.1038/sj.bmt.1704458] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The prognosis of inflammatory breast cancer (IBC) is poor. We evaluated clinical and biopathological characteristics that could affect survival in 74 women with nonmetastatic IBC consecutively treated in our institution between 1976 and 2000. Patients received primary anthracycline-based chemotherapy at conventional doses (n=20) or high-dose chemotherapy (HDC) with haematopoietic stem cell support (HSCS) (n=54). After chemotherapy, 84% of patients underwent mastectomy, 95% were given radiotherapy and 55% tamoxifen. Immunohistochemistry data (ER, PR, ERBB2, P53) on pre-chemotherapy specimens suggested strong differences between IBC and non-IBC. The rate of pathological complete response to chemotherapy was 26% (27% with HDC and 17% with conventional doses, not significant). No single factor was found predictive of response. With a median follow-up of 48 months after diagnosis, the 5-year projected disease-free survival (DFS) was 24% and overall survival (OS) 41%. In multivariate analysis, the strongest independent prognostic factor was the delivery of HDC. The 5-year DFS and OS of patients were respectively 28 and 50% with HDC and 15 and 18% with conventional chemotherapy. These results and comparisons with other series of patients suggest a role for HDC with HSCS as part of the therapeutic approach in IBC. Further prospective studies are required to confirm it.
Collapse
Affiliation(s)
- F Bertucci
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille Cedex 09, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Moffa AB, Tannheimer SL, Ethier SP. Transforming Potential of Alternatively Spliced Variants of Fibroblast Growth Factor Receptor 2 in Human Mammary Epithelial Cells. Mol Cancer Res 2004. [DOI: 10.1158/1541-7786.643.2.11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
A breast cancer cell line developed in our laboratory (SUM-52PE) has a 12-fold amplification and high-level overexpression of the oncogene fibroblast growth factor receptor 2 (FGFR2). Previously, nine different alternatively spliced FGFR2 variants were isolated from this cell line. Overexpression of two variants that differ only in their carboxyl termini (C1 and C3) has been successfully accomplished in the immortalized human mammary epithelial cell line H16N2. FGFR2 expression led to the activation of the mitogen-activated protein kinase and phosphatidylinositol 3-kinase signaling cascades. Phosphorylation of the adapter protein FGF receptor substrate 2 is much more robust in the cells expressing the C3 variant of FGFR2 compared with the C1 variant. H16N2 cells expressing the full-length FGFR2 with the C1 or C3 carboxyl terminus were tested for their ability to grow under epidermal growth factor (EGF)–independent conditions, in soft agar, and for their ability to invade naturally occurring basement membranes and compared with the parental SUM-52PE cell line. All three cell lines grew under EGF-independent conditions and all were inhibited by the FGFR family specific inhibitor PD173074. The full-length FGFR2-C1 and FGFR2-C3 variants grew robustly in soft agar similar to the parental cell line SUM-52PE. However, cells expressing the C3 variant formed large colonies in agar in both insulin-free and EGF-free medium, whereas the cells expressing the C1 variant required insulin for growth. Soft agar growth was also inhibited by PD173074. Because SUM-52PE was developed from a metastatic breast carcinoma, the FGFR2-overexpressing cell lines were assessed for their ability to invade sea urchin embryo cell membranes. H16N2 cells expressing the C1 carboxyl terminus failed to invade sea urchin embryo cell membranes. By contrast, FGFR2-C3-expressing cells were as invasive as the SUM-52 breast cancer cells and erbB-2-overexpressing H16N2 cells. These results indicate that FGFR2 is a transforming oncogene in human mammary epithelial cells when expressed to levels similar to that found in breast cancer cells with FGFR2 gene amplification. Furthermore, the results suggest that different splice variants have differing transforming activities and that signaling from variants expressing the C3 carboxyl terminus results in more autonomous signaling, cell growth, and invasion.
Collapse
Affiliation(s)
- Allison B. Moffa
- 1Cellular and Molecular Biology Graduate Program and
- 2Department of Radiation Oncology, Division of Radiation and Cancer Biology, University of Michigan Health System, Ann Arbor, Michigan
| | - Stacey L. Tannheimer
- 2Department of Radiation Oncology, Division of Radiation and Cancer Biology, University of Michigan Health System, Ann Arbor, Michigan
| | - Stephen P. Ethier
- 1Cellular and Molecular Biology Graduate Program and
- 2Department of Radiation Oncology, Division of Radiation and Cancer Biology, University of Michigan Health System, Ann Arbor, Michigan
| |
Collapse
|
18
|
Keohavong P, Gao WM, Mady HH, Kanbour-Shakir A, Melhem MF. Analysis of p53 mutations in cells taken from paraffin-embedded tissue sections of ductal carcinoma in situ and atypical ductal hyperplasia of the breast. Cancer Lett 2004; 212:121-30. [PMID: 15246568 DOI: 10.1016/j.canlet.2004.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2003] [Revised: 03/05/2004] [Accepted: 03/08/2004] [Indexed: 02/09/2023]
Abstract
Mutations in the p53 tumor suppressor gene are frequent in breast tumors but the implication of p53 mutations in breast cancer development remains poorly understood. In this study, we applied laser capture microdissection (LCM) microscope to histologically review and sample cells from paraffin-embedded breast tissue sections obtained from six cases of ductal carcinoma in situ (DCIS) and ten cases of atypical ductal hyperplasia (ADH). p53 mutations were detected, using single stranded conformational polymorphism (SSCP) and sequencing, in cell samples of three cases with DCIS and five cases with ADH. p53 mutations are therefore present in DCIS and ADH of the breast, considered as pre-malignant precursors to breast cancer, and some of them may represent early events in breast cancer development.
Collapse
Affiliation(s)
- Phouthone Keohavong
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, 3343 Forbes Avenue, Pittsburgh, PA 15260, USA.
| | | | | | | | | |
Collapse
|
19
|
Do TN, Rosal RV, Drew L, Raffo AJ, Michl J, Pincus MR, Friedman FK, Petrylak DP, Cassai N, Szmulewicz J, Sidhu G, Fine RL, Brandt-Rauf PW. Preferential induction of necrosis in human breast cancer cells by a p53 peptide derived from the MDM2 binding site. Oncogene 2003; 22:1431-44. [PMID: 12629507 DOI: 10.1038/sj.onc.1206258] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
p53 is the most frequently altered gene in human cancer and therefore represents an ideal target for cancer therapy. Several amino terminal p53-derived synthetic peptides were tested for their antiproliferative effects on breast cancer cell lines MDA-MB-468 (mutant p53), MCF-7 (overexpressed wild-type p53), and MDA-MB-157 (null p53). p53(15)Ant peptide representing the majority of the mouse double minute clone 2 binding site on p53 (amino acids 12-26) fused to the Drosophila carrier protein Antennapedia was the most effective. p53(15)Ant peptide induced rapid, nonapoptotic cell death resembling necrosis in all breast cancer cells; however, minimal cytotoxicity was observed in the nonmalignant breast epithelial cells MCF-10-2A and MCF-10F. Bioinformatic/biophysical analysis utilizing hydrophobic moment and secondary structure predictions as well as circular dichroism spectroscopy revealed an alpha-helical hydrophobic peptide structure with membrane disruptive potential. Based on these findings, p53(15)Ant peptide may be a novel peptide cancer therapeutic because it induces necrotic cell death and not apoptosis, which is uncommon in traditional cancer therapy.
Collapse
Affiliation(s)
- Tamara N Do
- Experimental Therapeutics Program, Division of Medical Oncology, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gnant MF, Dubsky PC, Stift A, Friedl J, Steger GG, Jakesz R, Zielinski CC. Future Directions in the Treatment of Breast Cancer – Cellular Interventions Using Dendritic Cells. Breast Cancer Res Treat 2003. [DOI: 10.1023/a:1026345612962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
21
|
Wen SF, Mahavni V, Quijano E, Shinoda J, Grace M, Musco-Hobkinson ML, Yang TY, Chen Y, Runnenbaum I, Horowitz J, Maneval D, Hutchins B, Buller R. Assessment of p53 gene transfer and biological activities in a clinical study of adenovirus-p53 gene therapy for recurrent ovarian cancer. Cancer Gene Ther 2003; 10:224-38. [PMID: 12637944 DOI: 10.1038/sj.cgt.7700562] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A cohort study was designed to evaluate the efficiency of gene transfer and whether biological activity from the expressed therapeutic gene resulted after administration of a recombinant adenovirus containing the human wild-type p53 (p53(wt)) gene (rAd-p53 SCH 58500). The cohort study was conducted in five trial subjects with recurrent ovarian cancer. Each trial subject received multiple cycles of rAd-p53 SCH 58500, each cycle comprised of doses of 7.5 x 10(13) particles on each of five consecutive days. Subjects were treated with rAd-p53 SCH 58500 alone during Cycle 1 and in combination with gemcitabine during the subsequent cycles. Both tumor biopsies and peritoneal aspirates were collected and evaluated for gene transfer and evidence of the biological activities of the expressed p53(wt) gene. Using quantitative PCR and RT-PCR, and in situ PCR, gene transfer and expression were documented in tumor biopsies (four of five patients) collected from Cycle 1. Furthermore, upregulation of p21/WAF1, bax and mdm-2, and downregulation of survivin were observed in these same tumor biopsy samples, suggesting that intraperitoneal administration of rAd-p53 SCH 58500 leads to detectable p53 biological activity in target tumor tissue. In addition, gene transfer and its expression were observed in cells obtained from peritoneal aspirates. These fluids were mainly comprised of polymorphonuclear neutrophils, indicating that successful gene transfer can be achieved by multiple cycle intraperitoneal administration of recombinant adenovirus.
Collapse
Affiliation(s)
- Shu Fen Wen
- Canji, Inc., San Diego, California 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Cell-cycle dysregulation is a hallmark of tumor cells. The ability of normal cells to undergo cell-cycle arrest after damage to DNA is crucial for the maintenance of genomic integrity. The biochemical pathways that stop the cell cycle in response to cellular stressors are called checkpoints. Defective checkpoint function results in genetic modifications that contribute to tumorigenesis. The regulation of checkpoint signaling also has important clinical implications because the abrogation of checkpoint function can alter the sensitivity of tumor cells to chemotherapeutics. Here, we provide an overview of the mechanisms that regulate the cell cycle, current anticancer therapies that target checkpoint signaling pathways, and strategies for the development of novel chemotherapeutic agents.
Collapse
Affiliation(s)
- Zoe A Stewart
- Department of Biochemistry, Center in Molecular Toxicology and the Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
23
|
Cengiz-Boduroglu E, Irkkan C, Bilir G. Is Nothingham Prognostic Index correlated with apoptosis and p53 expression in infiltrating ductal carcinoma of the breast? Pathol Oncol Res 2003; 9:100-3. [PMID: 12858214 DOI: 10.1007/bf03033752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2003] [Accepted: 06/20/2003] [Indexed: 11/25/2022]
Abstract
The role of p53 as a prognostic factor is not clear. P53 named as "guardian of the genome" plays an important role in many intracellular regulatory systems, one of which is apoptosis, having an impact on tumor kinetics. A retrospective study was undertaken to assess the relationship of the Nothingham Prognostic Index (NPI) to p53 expression and apoptotic cell counts. To conduct the study, 160 successive cases of infiltrating ductal carcinoma of the breast were included. P53 was assessed on AP-AAP stained sections. Apoptotic cell counting (ACC) was done on the HE stained routine sections in 10 HPFs. Clinical data were derived from the hospital files. Apoptotic cell counts were higher in the p53 positive group but the difference was not significant (p=0.079). P53 positivity was found to be related to the disease-free survival (DFS) (p=0.008). NPI was significantly higher in apoptotic cell containing group (p=0.006). There was a positive linear correlation between ACC and NPI scores (p=0.004). This correlation was not present between apoptosis and disease free survival. P53 expression was found to be related with DFS but not with the NPI which is a score composed of the best prognostic indicators known today. In contrast to this, ACC was found to be closely and linearly associated to the known prognostic factors. This may suggest that the apoptotic cell counts done on routine sections may be used as a part of prognosis assessment in infiltrating ductal carcinoma.
Collapse
|
24
|
Vergote D, Cren-Olivé C, Chopin V, Toillon RA, Rolando C, Hondermarck H, Le Bourhis X. (-)-Epigallocatechin (EGC) of green tea induces apoptosis of human breast cancer cells but not of their normal counterparts. Breast Cancer Res Treat 2002; 76:195-201. [PMID: 12462380 DOI: 10.1023/a:1020833410523] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
(-)-Epigallocatechin (EGC), one of green tea polyphenols, has been shown to inhibit growth of cancer cells. However its mechanism of action is poorly known. We show here that EGC strongly inhibited the growth of breast cancer cell lines (MCF-7 and MDA-MB-231) but not that of normal breast epithelial cells. The inhibition of breast cancer cell growth was due to an induction of apoptosis, without any change in cell cycle progression. MCF-7 cells are known to express a wild-type p53 whereas MDA-MB-231 cells express a mutated p53. The fact that EGC induced apoptosis in both these cell lines suggests that the EGC-triggered apoptosis is independent of p53 status. Moreover, neutralizing antibodies against the death receptor Fas and inhibitors of caspases, such as caspase-8 and -10, efficiently inhibited the EGC-triggered apoptosis. In addition, immunoblotting revealed that EGC treatment was correlated with a decrease in Bcl-2 and an increase in Bax level. These results suggest that EGC-triggered apoptosis in breast cancer cells requires Fas signaling.
Collapse
Affiliation(s)
- David Vergote
- Laboratoire de Biologic du Développement (UPRES-EA 1033), Université des Sciences et Technologies de Lille, Villeneuve d'Ascq, France
| | | | | | | | | | | | | |
Collapse
|
25
|
Qin C, Nguyen T, Stewart J, Samudio I, Burghardt R, Safe S. Estrogen up-regulation of p53 gene expression in MCF-7 breast cancer cells is mediated by calmodulin kinase IV-dependent activation of a nuclear factor kappaB/CCAAT-binding transcription factor-1 complex. Mol Endocrinol 2002; 16:1793-809. [PMID: 12145335 DOI: 10.1210/me.2002-0006] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
This study investigates the mechanism of hormonal regulation of p53 gene expression in MCF-7 human breast cancer cells. 17beta-Estradiol (E2) induced a 2-fold increase in p53 mRNA levels and a 2- to 3-fold increase in p53 protein. Analysis of the p53 gene promoter has identified a minimal E2-responsive region at -106 to -40, and mutation/deletion analysis of the promoter showed that motifs that bind CCAAT-binding transcription factor-1 (CTF-1) and nuclear factor kappaB (NFkappaB) proteins are required for hormone responsiveness. The p65 subunit of NFkappaB was identified in both nuclear and cytosolic fractions of untreated MCF-7 cells; however, formation of the nuclear NFkappaB complex was E2 independent. Hormonal activation of constructs containing p53 promoter inserts (-106 to -40) and the GAL4-p65 fusion proteins was inhibited by the intracellular Ca2+ ion chelator EGTA-AM and Ca2+/calmodulin-dependent protein kinase (CaMK) inhibitor KN-93. Constitutively active CaMKIV but not CaMKI activated p65, and treatment of MCF-7 cells with E2 induced phosphorylation of CaMKIV but not CaMKI. The results indicate that hormonal activation of p53 though nongenomic pathways was CaMKIV-dependent and involved cooperative p65-CTF-1 interactions.
Collapse
Affiliation(s)
- Chunhua Qin
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, Texas 77843-4466, USA
| | | | | | | | | | | |
Collapse
|
26
|
Bodey B. The significance of immunohistochemistry in the diagnosis and therapy of neoplasms. Expert Opin Biol Ther 2002; 2:371-93. [PMID: 11955276 DOI: 10.1517/14712598.2.4.371] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This review article details the diagnostical significance of immunohistochemistry, which has developed during the last quarter of the century. Certainly, the advancement of monoclonal antibody technology has been of great significance in assuring the place of immunohistochemistry in the modern accurate microscopic diagnosis of human neoplasms, as a method of choice in histopathology. The fact still remains that in order to properly assess any immunohistochemical reactivity used for differential diagnostic purposes, the target cells have to be identified as neoplastically transformed cells by routine histopathological techniques. Selected groups of target molecules of great significance in cancer biology are discussed. The discovery of neoplasm-associated antigens has not only made the more accurate diagnosis of human cancer feasible but has also shed light on the extensive immunophenotypical heterogeneity of even the most closely linked human malignancies. The identification of disseminated neoplastically transformed cells by immunohistochemistry has allowed for a clearer picture of cancer invasion and metastasis, as well as the evolution of the tumour cell associated immunophenotype towards increased malignancy. Some possibilities of neoplasm-associated antigen targeted, receptor-directed immunotherapy are discussed and reviewed in this manuscript. Future antineoplastic therapeutical approaches should see the inclusion of a variety of immunotherapies, in the form of an individualised 'cocktail' specific for the particular immunophenotypical pattern associated with each individual patient's neoplastic disease.
Collapse
Affiliation(s)
- Bela Bodey
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
27
|
Deb D, Scian M, Roth KE, Li W, Keiger J, Chakraborti AS, Deb SP, Deb S. Hetero-oligomerization does not compromise 'gain of function' of tumor-derived p53 mutants. Oncogene 2002; 21:176-89. [PMID: 11803461 DOI: 10.1038/sj.onc.1205035] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2001] [Revised: 09/14/2001] [Accepted: 10/09/2001] [Indexed: 01/10/2023]
Abstract
Tumor-derived p53 mutants activate transcription from promoters of various growth-related genes. We tested whether this transactivation function of the mutant protein is sufficient to induce tumorigenesis ('gain of function'). Tumor-derived mutant p53-281G transactivates the promoters of human epidermal growth factor receptor (EGFR) and human multiple drug resistance gene (MDR-1). To determine whether the C-terminal domain functions only as an oligomerization domain in mutant p53-mediated transactivation, we have replaced the tetramerization domain of p53 by a heterologous tetramerization domain; although this mutant protein formed tetramers in solution, it failed to transactivate significantly. Therefore, for successful mutant p53-mediated transactivation, sequences near the C-terminus of mutant p53 are required to perform functions in addition to tetramerization. We also demonstrate that co-expression of a deletion mutant of p53 (p53 del 1-293), which retains the p53 oligomerization domain, inhibits this transactivation. p53 del 1-293 co-immunoprecipitates with p53-281G suggesting that hetero-oligomers of p53-281G and p53 del 1-293 are defective in transactivation. We also show that a cell line stably transfected with p53-281G expresses higher levels of endogenous NF-kappaB and proliferating cell nuclear antigen (PCNA) compared to that transfected with vector alone. On co-expression, p53 del 1-293 lowered the levels of NF-kappaB and PCNA in p53-281G-expressing cells. However, on co-expression, p53 del 1-293 did not inhibit the tumorigenicity and colony forming ability of p53-281G expressing cells. Our earlier work showed that a deletion of the C-terminal sequences of p53-281G overlapping the oligomerization domain obliterates 'gain of function'. Taken together, the above information suggests that the C-terminal sequences have some critical role in 'gain of function' in addition to transactivation.
Collapse
Affiliation(s)
- Debabrita Deb
- Department of Biochemistry and Molecular Biophysics, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia, VA 23298, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Hirai M, LaFace D, Robinson S, Kelsey L, Johnson R, Wen SF, Warkentin P, Mills K, Vaillancourt M, Chavez J, Leutzinger C, Sumegi J, Neugebauer S, Lehman J, Talmadge C, Maneval D, Talmadge J. Ex vivo purging by adenoviral p53 gene therapy does not affect NOD-SCID repopulating activity of human CD34+ cells. Cancer Gene Ther 2001; 8:936-47. [PMID: 11781656 DOI: 10.1038/sj.cgt.7700390] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2001] [Indexed: 11/09/2022]
Abstract
Co-incubation of a replication-deficient, recombinant adenovirus carrying the wild-type p53 gene (rAd-p53) and hematopoietic stem cell (HSC) products from patients with breast cancer can significantly reduce tumor cell contamination. Whereas this approach provides a powerful tumor cell purging strategy, potential detrimental effects on the HSC population have not been investigated. The ability of human HSC to reconstitute hematopoiesis in severe combined immunodeficient (SCID) mice and to undergo secondary transplantation provides the only nonclinical measure of self-renewing, stem cell function. The objective of this study was to investigate whether co-incubation with rAd-p53 compromised the SCID repopulating activity (SRA) of HSC. Granulocyte colony-stimulating factor-mobilized human CD34+ cells were co-cultured with rAd-p53 at our targeted clinical dose, and the ability of these cells to establish multilineage hematopoiesis in sublethally irradiated, nonobese diabetic (NOD)-SCID mice was investigated. The persistence of human cells in the mice was investigated by flow cytometry, granulocyte-macrophage colony-forming unit assay, and polymerase chain reaction of human Alu sequences. Further, limiting dilution analysis provided a quantitative comparison between the SRA of CD34+ cells co-incubated with rAd-p53 and control CD34+ cells (no rAd-p53 co-incubation). We conclude that co-incubation with rAd-p53 has little effect on the SRA of HSC.
Collapse
Affiliation(s)
- M Hirai
- University of Nebraska Medical Center, Omaha, Nebraska 68198-7660, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Systemic p53 Gene Therapy of Cancer with Immunolipoplexes Targeted by Anti-Transferrin Receptor scFv. Mol Med 2001. [DOI: 10.1007/bf03401962] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
30
|
Elenbaas B, Spirio L, Koerner F, Fleming MD, Zimonjic DB, Donaher JL, Popescu NC, Hahn WC, Weinberg RA. Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. Genes Dev 2001; 15:50-65. [PMID: 11156605 PMCID: PMC312602 DOI: 10.1101/gad.828901] [Citation(s) in RCA: 663] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A number of genetic mutations have been identified in human breast cancers, yet the specific combinations of mutations required in concert to form breast carcinoma cells remain unknown. One approach to identifying the genetic and biochemical alterations required for this process involves the transformation of primary human mammary epithelial cells (HMECs) to carcinoma cells through the introduction of specific genes. Here we show that introduction of three genes encoding the SV40 large-T antigen, the telomerase catalytic subunit, and an H-Ras oncoprotein into primary HMECs results in cells that form tumors when transplanted subcutaneously or into the mammary glands of immunocompromised mice. The tumorigenicity of these transformed cells was dependent on the level of ras oncogene expression. Interestingly, transformation of HMECs but not two other human cell types was associated with amplifications of the c-myc oncogene, which occurred during the in vitro growth of the cells. Tumors derived from the transformed HMECs were poorly differentiated carcinomas that infiltrated through adjacent tissue. When these cells were injected subcutaneously, tumors formed in only half of the injections and with an average latency of 7.5 weeks. Mixing the epithelial tumor cells with Matrigel or primary human mammary fibroblasts substantially increased the efficiency of tumor formation and decreased the latency of tumor formation, demonstrating a significant influence of the stromal microenvironment on tumorigenicity. Thus, these observations establish an experimental system for elucidating both the genetic and cell biological requirements for the development of breast cancer.
Collapse
Affiliation(s)
- B Elenbaas
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Flatt PM, Pietenpol JA. Mechanisms of cell-cycle checkpoints: at the crossroads of carcinogenesis and drug discovery. Drug Metab Rev 2000; 32:283-305. [PMID: 11139130 DOI: 10.1081/dmr-100102335] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Human tumors arise from multiple genetic changes that gradually transform growth-limited cells into highly invasive cells that are unresponsive to growth controls. The genetic evolution of normal cells into cancer cells is largely determined by the fidelity of DNA replication, repair, and division. Cell-cycle arrest in response to stress is integral to the maintenance of genomic integrity. The control mechanisms that restrain cell-cycle transition or induce apoptotic signaling pathways after cell stress are known as cell-cycle checkpoints. This review will focus on the mechanisms of cell-cycle checkpoint pathways and how different components of these pathways are frequently altered in the genesis of human tumors. As our knowledge of cell-cycle regulation and checkpoints increases, so will our understanding of how xenobiotic agents can affect these processes to either initiate or inhibit tumorigenesis.
Collapse
Affiliation(s)
- P M Flatt
- Department of Biochemistry, Center in Molecular Toxicology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
32
|
Abstract
Chromosome instability (a high frequency of chromosomal loss and gain and genome doubling, often referred to as karyotypic instability) is one of the major characteristics of cancer cells. It facilitates carcinogenesis by increasing the chance of specific mutations responsible for malignant phenotypes. Chromosome instability in most cases reflects the occurrence of defective mitosis, including unequal distribution of chromosomes to daughter cells and failure to undergo cytokinesis, which leads to generation of aneuploid cells. Both in vivo and in vitro, chromosome instability has been shown to correlate with loss or mutation of the p53 tumor suppressor protein, the product of one of the most frequently mutated genes in cancer. The major function of p53 is to prevent cells from proceeding through the cell cycle when cells experience stress, insults, or errors that disturb the preprogrammed cell cycle progression. During the last several years, significant advances have been made in understanding how p53 is involved in the regulation of mitosis and how loss or mutation of p53 affects mitotic fidelity, which will be the subject of this review.
Collapse
Affiliation(s)
- P Tarapore
- Department of Cell Biology, University of Cincinnati College of Medicine, Ohio, USA
| | | |
Collapse
|
33
|
Gion M, Boracchi P, Dittadi R, Biganzoli E, Peloso L, Gatti C, Paccagnella A, Rosabian A, Vinante O, Meo S. Quantitative measurement of soluble cytokeratin fragments in tissue cytosol of 599 node negative breast cancer patients: a prognostic marker possibly associated with apoptosis. Breast Cancer Res Treat 2000; 59:211-21. [PMID: 10832591 DOI: 10.1023/a:1006318112776] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Apoptosis is associated with caspase-mediated proteolysis of Type I (K18 and K19) cytokeratins. We previously showed a positive association between the levels of tissue polypeptide antigen (TPA), that recognizes cytokeratins K8, K18, and K19 fragments, and induced apoptosis in breast cancer cell lines. The aim of the present study was to evaluate the interrelationships between TPA, steroid receptors, and p53, and their joint prognostic role in node-negative breast cancer patients not treated with adjuvant therapies. Age and pT were also considered since they are known prognostic factors. Five hundred and ninety-nine cases with N- breast cancer were evaluated (median follow-up: 60 months). TPA was measured by an immunoradiometric assay and p53 by an immunochemiluminescent assay in tumor cytosol. Multiple correspondence analysis was used to study the associations among variables. Their prognostic role (univariate analysis) and their joint effect (multivariate analysis) on RFS were investigated with Cox regression models. TPA showed a direct association with ER and PgR. Higher p53 values were weakly associated to low values of ER, PgR, and TPA. Younger age was related to low and intermediate values of ER and PgR and to low p53 values, while older age was related to high values of ER. Multivariate analysis showed a significant prognostic impact for pT, age, ER, and TPA. Among the interactions considered clinically relevant, only that between ER and age was found. RFS estimated values were poorer in cases with lower than in those with higher TPA values, both in patients expected to have a poor (pT2, young age, low ER) and a better prognosis (pT1, older age, high ER). From the findings of the present study we can draw the following conclusions: The relationship of TPA with prognosis gives an additional contribution to pT, age, and steroid receptors in N- breast cancer; TPA may be considered the first marker of apoptosis measured with a fully standardized quantitative method in tumor cytosol and could be evaluated in prognostic indexes including markers related to different biological mechanisms.
Collapse
Affiliation(s)
- M Gion
- Centro Regionale per lo Studio degli Indicatori Biochimici di Tumore, Ospedale Civile, Venezia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lê MG, Mathieu MC, Douc-Rasy S, Le Bihan ML, Adb El All H, Spielmann M, Riou G. c-myc, p53 and bcl-2, apoptosis-related genes in infiltrating breast carcinomas: evidence of a link between bcl-2 protein over-expression and a lower risk of metastasis and death in operable patients. Int J Cancer 1999; 84:562-7. [PMID: 10567899 DOI: 10.1002/(sici)1097-0215(19991222)84:6<562::aid-ijc4>3.0.co;2-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Apoptosis is an important physiological process controlled by multiple genes, including c-myc, p53 and bcl-2, and its inhibition may lead to the development of human cancers. In this study, we analyzed expression of the c-myc gene using Northern blot and of the p53 and bcl-2 proteins by immuno-histochemistry in 175 breast tumor specimens obtained from patients with operable breast cancer. We evaluated the relation between expression of these 3 genes and (i) the main usual prognostic factors (tumor size, histo-prognostic grade, hormone receptors and number of positive nodes); (ii) the risk of death and relapse, taking into account these 4 factors, after a mean period of follow-up of 9.5 years (SD 2 years). Over-expression of c-myc, p53 and bcl-2 was observed in 35%, 23% and 63% of tumors, respectively. Over-expression of c-myc was strongly linked to the number of positive nodes (p = 0.0005). p53 protein expression was associated with both high-grade (p = 0.0001) and hormone receptor-negative (p = 0.0001) tumors. In contrast, bcl-2 protein over-expression was associated with the main favorable prognostic factors and, more particularly, with hormone receptor-positive tumors (p = 0.0001). Multivariate analysis, using the Cox model, showed that only 2 factors were independently linked to the risk of death: number of positive nodes, which increased the risk (p = 0.0001), and bcl-2 protein over-expression, which decreased the risk (p = 0.008). When bcl-2 over-expression was studied in relation to nodal status, hormone receptor status and chemo- and hormone therapy, no significant difference was observed between different subgroups of patients. bcl-2 expression was also associated with a significantly lower risk of distant metastasis (p = 0.04). In conclusion, bcl-2 expression characterizes a particular phenotype of breast cancer with a favorable prognosis, and it may therefore be used as a marker of long-term survival. Int. J. Cancer (Pred. Oncol.) 84:562-567, 1999.
Collapse
Affiliation(s)
- M G Lê
- Institut National de la Santé et de la Recherche Médicale (INSERM-U521), Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Meng L, Lin L, Zhang H, Nassiri M, Morales AR, Nadji M. Multiple mutations of the p53 gene in human mammary carcinoma. Mutat Res 1999; 435:263-9. [PMID: 10606817 DOI: 10.1016/s0921-8777(99)00053-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Alteration of the p53 tumor suppressor gene is the most common genetic abnormality in human cancer. In breast cancer, depending on the stage of disease and method of detection, mutation rates of 25-60% have been observed. Multiple mutations of p53 gene in the same tumor however, are rarely reported. In this study we explored the frequency of multiple mutations of p53 gene in mammary carcinoma in a cohort of south Florida patients. Three hundred eighty-four cases of primary breast cancer diagnosed between 1984 and 1986 at the University of Miami, Jackson Medical Center were subjects of this study. Sequence analysis of exons 5 through 8 of p53 was performed on cloned PCR-amplified DNA of formalin-fixed, paraffin-embedded tumors. Two hundred thirty-four of 384 breast cancers (61%) had p53 mutation. Of those, 36 tumors showed more than one mutation; 31 tumors had two mutations, three showed three, one tumor had five mutations, and one case carried six mutations. The majority of mutations were missense (43) followed by silent (35); and most occurred within a single exon. Our study suggests that multiple mutations of p53 suppressor gene in breast cancer are more common than currently believed.
Collapse
Affiliation(s)
- L Meng
- Department of Pathology, University of Miami, Jackson Medical Center, UM/JMC, P.O. Box 016960, Miami, FL 33101, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Most human breast tumors arise from multiple genetic changes which gradually transform differentiated and growth-limited cells into highly invasive cells that are unresponsive to growth controls. The genetic evolution of normal breast cells into cancer cells is largely determined by the fidelity of DNA replication, repair, and division. Cell cycle arrest in response to DNA damage is an important part of the mechanism used to maintain genomic integrity. The control mechanisms that restrain cell cycle transition after DNA damage are known as cell cycle checkpoints. This review will focus on cell cycle checkpoint signaling pathways commonly mutated in human breast tumors and suggest how different components of these checkpoint pathways offer the potential for chemotherapeutic intervention.
Collapse
Affiliation(s)
- Z A Stewart
- Department of Biochemistry, Center in Molecular Toxicology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
37
|
Würl P, Meye A, Lautenschläger C, Schmidt H, Bache M, Kalthoff H, Schönfelder M, Rath FW, Taubert H. Clinical relevance of pRb and p53 co-overexpression in soft tissue sarcomas. Cancer Lett 1999; 139:159-65. [PMID: 10395173 DOI: 10.1016/s0304-3835(99)00034-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The goal of this study was to examine the relationship between immunohistochemical pRb detectability and p53 overexpression in 198 soft tissue sarcomas (STS) with regard to its clinical relevance. Distinct pRb detectability multivariately shows a correlation to survival rate (relative risk (RR)=1.59, P=0.037). p53 positivity was also multivariately correlated to poor prognosis (RR=2.17, P=0.0014). Stratification of pRb staining to p53 results shows a prognostical graduation. Patients with negativity for both proteins have the most favorable prognosis (projected 5-year survival rate (psr)=54.5%). In contrast to this, positivity for both antibodies has the highest risk (RR=2.48, P=0.02) and the poorest prognosis (psr=17.4%). To conclude, these results explain that the clinical relevance of immunohistochemical pRb positivity in STS is connected with p53 in the form of having an increasing effect on the known prognostic relevance of p53 overexpression.
Collapse
Affiliation(s)
- P Würl
- Surgical Clinic 1, University of Leipzig, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Carroll PE, Okuda M, Horn HF, Biddinger P, Stambrook PJ, Gleich LL, Li YQ, Tarapore P, Fukasawa K. Centrosome hyperamplification in human cancer: chromosome instability induced by p53 mutation and/or Mdm2 overexpression. Oncogene 1999; 18:1935-44. [PMID: 10208415 DOI: 10.1038/sj.onc.1202515] [Citation(s) in RCA: 214] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We have previously reported that loss of p53 tumor suppressor protein results in centrosome hyperamplification, which leads to aberrant mitosis and chromosome instability. Since p53 is either deleted or mutated in human cancers at a high frequency, we investigated whether human cancers showed centrosome hyperamplification. Screening of advanced stage breast ductal carcinomas and squamous cell carcinomas of the head and neck (SCCHN) revealed that centrosome hyperamplification is frequent in both tumor types. Moreover, through the analyses of p53 in SCCHN samples by direct sequencing and by loss-of-heterozygosity test, we found that p53 mutations correlated with occurrence of centrosome hyperamplification. However, in some cases, we observed centrosome hyperamplification in tumors that retained wild-type p53. These tumors contained high levels of Mdm2. Since Mdm2 can inactivate p53 through physical association, we investigated whether Mdm2 overexpression induced centrosome hyperamplification. We found that Mdm2 overexpression, like loss of p53, induced centrosome hyperamplification and chromosome instability in cultured cells.
Collapse
Affiliation(s)
- P E Carroll
- Department of Cell Biology, University of Cincinnati College of Medicine, Ohio 45267-0521, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Furth PA. Apoptosis and the Development of Breast Cancer. Breast Cancer 1999. [DOI: 10.1007/978-1-59259-456-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
40
|
Koifman RJ, Koifman S, Vieira RJ. Familial aggregation of breast/ovarian cancer: age of onset along subsequent generations in Brazil. CAD SAUDE PUBLICA 1998; 14 Suppl 3:181-5. [PMID: 9819477 DOI: 10.1590/s0102-311x1998000700019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Antecedents of family aggregation of breast and ovarian cancer are observed in only 5-8% of all breast cancer cases. Nevertheless, this variable displays one of the highest risk ratios associated to breast cancer outcome. Despite recent identification of genetic mutations associated with family aggregation of these tumors, mainly at BRCA1 and BRCA2 genes, knowledge on the interaction between environmental agents in these families remains quite unclear. In this paper we ascertained the correlation among ages of the onset of breast/ovarian cancer in 260 Brazilian families with those cancer aggregations. Further we estimated the median age of the onset of breast cancer among four generations. We observed that the higher the number of family cancer cases, the highest is the correlation of ages for the onset of breast cancer. We also observed a 8-10 year decline in the mean age-of-onset of breast/ovarian cancer from one generation to another in the studied families. If these results could be confirmed elsewhere, we believe that the hypothesis of interaction between environmental risks factors in families indeed showing breast/ovarian cancer aggregation is reinforced.
Collapse
Affiliation(s)
- R J Koifman
- Departamento de Epidemiologia, Escola Nacional de Saúde Pública, Fundação Oswaldo Cruz. Rua Leopoldo Bulhões 1480, Rio de Janeiro, RJ 21041-210, Brasil.
| | | | | |
Collapse
|
41
|
Trepel M, Groscurth P, Malipiero U, Gulbins E, Dichgans J, Weller M. Chemosensitivity of human malignant glioma: modulation by p53 gene transfer. J Neurooncol 1998; 39:19-32. [PMID: 9760067 DOI: 10.1023/a:1005910323338] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Loss of wild-type p53 activity is one of the most common molecular abnormalities in human cancers including malignant gliomas. The p53 status is also thought to modulate sensitivity to irradiation and chemotherapy. Here, we studied the effect of a p53 gene transfer on the chemosensitivity of three human glioma cell lines with different endogenous p53 status (LN-229, wild-type; LN-18, mutant; LN-308, deleted), using the murine temperature-sensitive p53 val135 mutant. Expression of mutant p53 enhanced proliferation of LN-308 cells but reduced proliferation in the other cell lines. Expression of wild-type p53 caused reversible growth arrest of all cell lines but failed to induce apoptosis. Growth arrest induced by wild-type p53 was associated with strong induction of p21 expression. Strong induction of BAX expression and loss of BCL-2 expression, which are associated with p53-dependent apoptosis rather than growth arrest, were not observed. Wild-type p53 failed to sensitize glioma cells to cytotoxic drugs including BCNU, cytarabine, doxorubicin, teniposide and vincristine. The combined effects of wild-type p53 gene transfer and drug treatment were less than additive rather than synergistic, suggesting that the intracellular cascades activated by p53 and chemotherapy are redundant. Unexpectedly, forced expression of mutant p53 modulated drug sensitivity in that it enhanced the toxicity of some drugs but attenuated the effects of others. These effects may represent a dominant negative effect of mutant p53 in LN-229 cells which have wild-type p53 activity but must be considered a gain of function-type effect in the other two cell lines which have no wild-type p53 activity. Importantly, no clear-cut pattern emerged among the three cell lines studied. We conclude that somatic gene therapy based on the reintroduction of p53 will limit the proliferation of human malignant glioma cells but is unlikely to induce clinically relevant sensitization to chemotherapy in these tumors.
Collapse
Affiliation(s)
- M Trepel
- Department of Neurology, University of Tübingen, Medical School, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Medina D, Stephens LC, Bonilla PJ, Hollmann CA, Schwahn D, Kuperwasser C, Jerry DJ, Butel JS, Meyn RE. Radiation-induced tumorigenesis in preneoplastic mouse mammary glands in vivo: significance of p53 status and apoptosis. Mol Carcinog 1998; 22:199-207. [PMID: 9688146 DOI: 10.1002/(sici)1098-2744(199807)22:3<199::aid-mc8>3.0.co;2-g] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In mouse mammary tumorigenesis, p53 mutations facilitate tumorigenesis in concert with other oncogenic alterations. Ionizing radiation enhances tumorigenesis in preneoplastic mammary outgrowth lines and induces p53-dependent apoptosis. We asked if normal p53 function modulates radiation-induced tumorigenesis in preneoplastic mammary lesions by affecting the apoptotic pathway of cell deletion. Three different hyperplastic outgrowth lines were compared. Outgrowth line D1 overexpressed wild-type p53 and responded to irradiation with enhanced tumorigenicity but no induction of apoptosis. Outgrowth line TM12 exhibited normal wild-type p53 expression and responded to irradiation with no alteration in tumorigenicity but with a marked increase in apoptosis. Outgrowth line TM2L also exhibited normal wild-type p53 expression and responded to irradiation with a marked enhancement in both tumorigenicity and apoptosis. These results indicate that the two radiation-induced responses, apoptosis and tumorigenesis, are dissociable events in the mammary gland. Furthermore, radiation-induced tumorigenicity was not abrogated by either enhanced wild-type p53 expression or a robust apoptotic response. The radiation dose of 5 Gy most likely induces multiple genetic alterations in surviving cells, including genomic instability, and this may account for the tumorigenicity. Future experiments will examine lower doses of irradiation that still induce a significant apoptotic response but significantly less genomic instability.
Collapse
Affiliation(s)
- D Medina
- Department of Cell Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Affiliation(s)
- V Chiarugi
- Laboratory of Molecular Biology, University of Florence, Italy
| | | | | |
Collapse
|
45
|
Nielsen LL, Gurnani M, Syed J, Dell J, Hartman B, Cartwright M, Johnson RC. Recombinant E1-deleted adenovirus-mediated gene therapy for cancer: efficacy studies with p53 tumor suppressor gene and liver histology in tumor xenograft models. Hum Gene Ther 1998; 9:681-94. [PMID: 9551616 DOI: 10.1089/hum.1998.9.5-681] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Type 5 adenoviral (Ad) vectors have been the "vector-of-choice" for preclinical studies on p53 tumor suppressor gene therapy of cancer. Previous studies have examined the in vivo efficacy of p53 Ad when given intratumorally. However published information does little to guide clinicians in the design of intraperitoneal (i.p.) dosing trials for i.p. tumors, e.g., ovarian, or clinical trials using regional organ perfusion, e.g., for lung tumors. Therefore, we examined several parameters with special significance for these routes of administration. Lung metastases from p53mut MDA-MB-231 mammary xenografts were treated with therapeutic levels of intravenous buffer, beta-galactosidase (beta-Gal) Ad, or p53 Ad. Treatment with intravenous p53 Ad significantly reduced the number of metastases per lung and there was a dramatic reduction in the surface area occupied by these tumors as compared to control groups. Two types of i.p. tumor xenografts were used for preclinical modeling of i.p. gene therapy, the p53null SK-OV-3 ovarian and the p53mut DU-145 prostate human cancers. In a study examining the effect of different vehicle volumes on the efficacy of a constant drug dose, all mice treated with p53 Ad had reduced tumor burden compared to controls. Dosing volumes between 0.2 and 1 ml were equally effective and all were more effective than a dosing volume of 0.1 ml. However, reduced efficacy was observed when a volume of 1.5 ml was used. When the effect of dosing frequency on antitumor efficacy was examined, fractionated doses of p53 Ad had somewhat greater efficacy than fewer, bolus injections. One of the significant elements in the emerging toxicology associated with recombinant adenoviruses is the hepatocyte pathology caused by high systemic concentrations of adenovirus. For recombinant Ad used in this study, there was a pronounced dose-dependence for the liver response, with very high, repeated doses causing significant hepatocellular insult. Expression of cytoplasmic beta-Gal protein coincided with areas of greatest damage in mice treated with high doses of beta-Gal Ad. Ultrastructural examination of hepatocyte intranuclear inclusions revealed moderately electron-dense, tightly packed granular material interspersed with more electron-dense nuclear material. Human tumor xenografts, but not mouse tissues, expressed viral hexon protein. In summary, hepatic toxicity caused by high concentrations of recombinant adenovirus was observed in murine cancer models. However, therapeutic levels of p53 Ad could be achieved which had dramatic efficacy without significant pathology.
Collapse
Affiliation(s)
- L L Nielsen
- Tumor Biology Department, Schering-Plough Research Institute, Kenilworth, NJ 07033, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Moll UM, Schramm LM. p53--an acrobat in tumorigenesis. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 1998; 9:23-37. [PMID: 9488246 DOI: 10.1177/10454411980090010101] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The p53 tumor suppressor protein plays a central role in maintaining genomic integrity. It does so by occupying a nodal point in the DNA damage control pathway. When cells are subject to ionizing radiation or other mutagenic events, p53 mediates cell cycle arrest or programmed cell death (apoptosis). Furthermore, some evidence suggests that p53 plays a role in the recognition and repair of damaged DNA. Biochemically, p53 is a sequence-specific transcriptional stimulator and a non-specific transcriptional repressor but also engages in multiple protein-protein interactions. Conversely, disruption of the p53 response pathway strongly correlates with tumorigenesis. p53 is functionally inactivated by structural mutations, neutralization by viral products, and non-mutational cellular mechanisms in the majority of human cancers. p53-deficient mice have a highly penetrant tumor phenotype, with over 90% tumor incidence within nine months. In some cancers, direct physical evidence exists identifying the p53 gene as a target of known environmental carcinogens such as UV light and benzolalpyrene in cancers of the skin and lung. When p53 loss occurs, cells do not get repaired or eliminated but rather proceed to replicate damaged DNA, which results in more random mutations, gene amplifications, chromosomal re-arrangements, and aneuploidy. In some experimental models, loss of p53 confers resistance to anticancer therapy due to loss of apoptotic competence. The translational potential of these discoveries is beginning to be tested in novel p53-based therapies.
Collapse
Affiliation(s)
- U M Moll
- Department of Pathology, Health Sciences Center, State University of New York at Stony Brook, 11794-8691, USA
| | | |
Collapse
|
47
|
Iype, Michael, Verma, Iype. Development and characterization of new immortalized human breast cell lines. Cytotechnology 1998; 26:207-18. [PMID: 9618944 PMCID: PMC3449551 DOI: 10.1023/a:1007953429252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
New human breast cell lines were developed from metastatic breast cancer tissues and normal breast tissues. Primary cultures were initiated from cellular outgrowths of explanted tissues or from mechanically isolated cells in two serum-free media. Cell cultures derived from both cancer and normal tissues were immortalized with pRSV-T plasmid to generate permanent breast cell lines that exhibited an epithelial morphology. Cell lines generated in this study were characterized with respect to morphology, growth rate, karyotype, presence of specific genes, and the expression of epithelial and breast markers. The cell lines expressed the epithelial cell markers, cytokeratins 8 and 18, and retained the capacity to produce human milk fat globulin. They also express the BRCA-1, erbB2, and EGF receptor genes and possess the H-ras, K-ras, and p53 genes. Preliminary data showed that one of the new cancer cell lines was highly sensitive to the cytotoxic action of taxol. It is envisioned that the new breast cell lines will be useful as targets for identification of therapeutic agents against breast cancer and as models for carcinogenesis studies.
Collapse
Affiliation(s)
- Iype
- Biological Research Faculty and Facility, Inc., Ijamsville, Maryland 21754, USA
| | | | | | | |
Collapse
|
48
|
Fixation Time and Microwave Oven Irradiation Affect Immunocytochemical p53 Detection in Formalin-Fixed Paraffin Sections. ACTA ACUST UNITED AC 1998. [DOI: 10.1097/00022744-199809000-00006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Winthrop MD, DeNardo SJ, Muenzer JT, Chi SG, Gumerlock PH. p53-independent response of a human breast carcinoma xenograft to radioimmunotherapy. Cancer 1997. [DOI: 10.1002/(sici)1097-0142(19971215)80:12+<2529::aid-cncr27>3.0.co;2-d] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
50
|
Magnelli L, Ruggiero M, Chiarugi V. The old and the new in p53 functional regulation. BIOCHEMICAL AND MOLECULAR MEDICINE 1997; 62:3-10. [PMID: 9367792 DOI: 10.1006/bmme.1997.2616] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The gene termed p53 is one of the most extensively studied for the past 18 years and the amount of literature published on this gene reflects its relevance in the field of molecular oncology; thus, loss or mutation of this oncosuppressor gene is probably the molecular lesion most frequently observed in human tumors. The aim of this minireview is to report, discuss, and interpret some recent observations on this topic: (I) The relationship with the Ataxia-Telangectasia gene and with the signaling enzyme phosphatidylinositol 3-kinase (PI3K). (II) The relationship between DNA damage, p53, and sensitivity to anticancer therapies. (III) The gain of function caused by mutations that transform the oncosuppressor p53 gene into a dominant transforming oncogene and (IV) The phosphorylative regulation of p53 and its relationship with the mitogenic signaling cascade involving protein kinase C and tumor promoters.
Collapse
Affiliation(s)
- L Magnelli
- Laboratory of Molecular Biology at the Institute of General Pathology, University of Florence , Viale Morgagni 50, Florence, 50134, Italy
| | | | | |
Collapse
|