1
|
Feng FY, Zhang Y, Kothari V, Evans JR, Jackson WC, Chen W, Johnson SB, Luczak C, Wang S, Hamstra DA. MDM2 Inhibition Sensitizes Prostate Cancer Cells to Androgen Ablation and Radiotherapy in a p53-Dependent Manner. Neoplasia 2017; 18:213-22. [PMID: 27108384 PMCID: PMC4840291 DOI: 10.1016/j.neo.2016.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 01/27/2016] [Indexed: 11/23/2022]
Abstract
PURPOSE: Increased murine double minute 2 (MDM2) expression, independent of p53 status, is associated with increased cancer-specific mortality for men with prostate cancer treated with radiotherapy. We assessed MI-219, a small molecule inhibitor of MDM2 with improved pharmacokinetics over nutlin-3, for sensitization of prostate cancer cells to radiotherapy and androgen deprivation therapy, a standard treatment option for men with high-risk prostate cancer. EXPERIMENTAL DESIGN: The effect of MDM2 inhibition by MI-219 was assessed in vitro and in vivo with mouse xenograft models across multiple prostate cancer cell lines containing varying p53 functional status. RESULTS: MDM2 inhibition by MI-219 resulted in dose- and time-dependent p53 activation and decreased clonogenic cell survival after radiation in a p53-dependent manner. Mechanistically, radiosensitization following inhibition of MDM2 was largely the result of p53-dependent increases in apoptosis and DNA damage as evidenced by Annexin V flow cytometry and γ-H2AX foci immunofluorescence. Similarly, treatment with MI-219 enhanced response to antiandrogen therapy via a p53-dependent increase in apoptotic cell death. Lastly, triple therapy with radiation, androgen deprivation therapy, and MI-219 decreased xenograft tumor growth compared with any single- or double-agent treatment. CONCLUSION: MDM2 inhibition with MI-219 results in p53-dependent sensitization of prostate cancer cells to radiation, antiandrogen therapy, and the combination. These findings support MDM2 small molecule inhibitor therapy as a therapy intensification strategy to improve clinical outcomes in high-risk localized prostate cancer. TRANSLATIONAL RELEVANCE: The combination of radiotherapy and androgen deprivation therapy is a standard treatment option for men with high-risk prostate cancer. Despite improvements in outcomes when androgen deprivation therapy is added to radiation, men with high-risk prostate cancer have significant risk for disease recurrence, progression, and even death within the first 10 years following treatment. We demonstrate that treatment with MI-219 (an inhibitor of MDM2) results in prostate cancer cell sensitization to radiation and androgen deprivation therapy in vitro and in vivo. Triple therapy with MI-219, radiation, and androgen deprivation therapy dramatically decreased tumor growth compared with any single- or double-agent therapy. These findings provide evidence that inhibition of MDM2 is a viable means by which to enhance the efficacy of both radiation and androgen deprivation therapy and thereby improve outcomes in the treatment of prostate cancer. As such, further investigation is warranted to translate these findings to the clinical setting.
Collapse
Affiliation(s)
- Felix Y Feng
- Departments of Radiation Oncology, Urology, and Medicine, University of California at San Francisco, San Francisco, CA; Helen Diller Family Comprehensive Cancer Center; Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.
| | - Yu Zhang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Vishal Kothari
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Joseph R Evans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - William C Jackson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Wei Chen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Skyler B Johnson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Connor Luczak
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Shaomeng Wang
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA; Departments of Internal Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Daniel A Hamstra
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Shinohara ET, Lu B, Hallahan DE. The Use of Gene Therapy in Cancer Research and Treatment. Technol Cancer Res Treat 2016; 3:479-90. [PMID: 15453813 DOI: 10.1177/153303460400300509] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Gene therapy involves identifying a gene of interest and then manipulating the expression of this gene through a variety of techniques. Here we specifically address gene therapy's role in cancer research. This paper will encompass thoroughly investigated techniques such as cancer vaccines and suicide gene therapy and the latest advancements in and applications of these techniques. It will also cover newer techniques such as Antisense Oligonucleotides and small interfering RNAs and how these technologies are being developed and used. The use of gene therapy continues to expand in cancer research and has an integral role in the advancement of cancer treatment.
Collapse
Affiliation(s)
- E T Shinohara
- Department of Radiation Oncology, Vanderbilt University, 1301 22nd Avenue South, B-902, The Vanderbilt Clinic, Nashville, Tennessee 37232-5671, USA
| | | | | |
Collapse
|
3
|
Murine double minute 2 siRNA and wild-type p53 gene therapy enhances sensitivity of the SKOV3/DDP ovarian cancer cell line to cisplatin chemotherapy in vitro and in vivo. Cancer Lett 2014; 343:200-9. [DOI: 10.1016/j.canlet.2013.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/27/2013] [Accepted: 10/07/2013] [Indexed: 01/03/2023]
|
4
|
|
5
|
Jiang T, Zhou C, Gu J, Liu Y, Zhao L, Li W, Wang G, Li Y, Cai L. Enhanced therapeutic effect of cisplatin on the prostate cancer in tumor-bearing mice by transfecting the attenuated Salmonella carrying a plasmid co-expressing p53 gene and mdm2 siRNA. Cancer Lett 2013; 337:133-42. [PMID: 23726840 DOI: 10.1016/j.canlet.2013.05.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/17/2013] [Accepted: 05/23/2013] [Indexed: 12/12/2022]
Abstract
Prostate cancer urgently needs an efficient therapy. Here we demonstrated that cisplatin combined with gene therapy by transfecting the attenuated Salmonella that carry a plasmid containing p53 gene and MDM2 siRNA provided a super-synergistic effect on the inhibition of prostate cancer growth in vivo. This synergistic therapy was associated with the induction of apoptotic cell death with a decreased Bcl2 to Bax expression ratio and increased expression of cleaved caspase 3 and caspase 9 in the prostate cancer xenograft. These results indicate that cisplatin-chemotherapy in combination with targeting the MDM2/p53 axis is an attractive strategy to treat prostate cancer.
Collapse
Affiliation(s)
- Tao Jiang
- Cancer Center at the First Hospital of Jilin University, Changchun 130021, China
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Tran PT, Hales RK, Zeng J, Aziz K, Salih T, Gajula RP, Chettiar S, Gandhi N, Wild AT, Kumar R, Herman JM, Song DY, DeWeese TL. Tissue biomarkers for prostate cancer radiation therapy. Curr Mol Med 2012; 12:772-87. [PMID: 22292443 PMCID: PMC3412203 DOI: 10.2174/156652412800792589] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 11/10/2011] [Accepted: 12/20/2011] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the most common cancer and second leading cause of cancer deaths among men in the United States. Most men have localized disease diagnosed following an elevated serum prostate specific antigen test for cancer screening purposes. Standard treatment options consist of surgery or definitive radiation therapy directed by clinical factors that are organized into risk stratification groups. Current clinical risk stratification systems are still insufficient to differentiate lethal from indolent disease. Similarly, a subset of men in poor risk groups need to be identified for more aggressive treatment and enrollment into clinical trials. Furthermore, these clinical tools are very limited in revealing information about the biologic pathways driving these different disease phenotypes and do not offer insights for novel treatments which are needed in men with poor-risk disease. We believe molecular biomarkers may serve to bridge these inadequacies of traditional clinical factors opening the door for personalized treatment approaches that would allow tailoring of treatment options to maximize therapeutic outcome. We review the current state of prognostic and predictive tissue-based molecular biomarkers which can be used to direct localized prostate cancer treatment decisions, specifically those implicated with definitive and salvage radiation therapy.
Collapse
Affiliation(s)
- P T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, 1550 Orleans Street, CRB2, RM 406, Baltimore, MD 21231, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Suganthi M, Sangeetha G, Benson CS, Babu SD, Sathyavathy A, Ramadoss S, Ravi Sankar B. In vitro mechanisms involved in the regulation of cell survival by lithium chloride and IGF-1 in human hormone-dependent breast cancer cells (MCF-7). Toxicol Lett 2012; 214:182-91. [DOI: 10.1016/j.toxlet.2012.08.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/17/2012] [Accepted: 08/25/2012] [Indexed: 01/24/2023]
|
8
|
Ji K, Wang B, Shao YT, Zhang L, Liu YN, Shao C, Li XJ, Li X, Hu JD, Zhao XJ, Xu DQ, Li Y, Cai L. Synergistic Suppression of Prostatic Cancer Cells by Coexpression of BothMurine Double Minute 2Small Interfering RNA and Wild-Typep53Gene In Vitro and In Vivo. J Pharmacol Exp Ther 2011; 338:173-83. [DOI: 10.1124/jpet.111.180364] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
9
|
Udayakumar T, Shareef MM, Diaz DA, Ahmed MM, Pollack A. The E2F1/Rb and p53/MDM2 pathways in DNA repair and apoptosis: understanding the crosstalk to develop novel strategies for prostate cancer radiotherapy. Semin Radiat Oncol 2011; 20:258-66. [PMID: 20832018 DOI: 10.1016/j.semradonc.2010.05.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Both the p53- and E2F1-signaling pathways are defective in almost all types of tumors, suggesting very important roles for their signaling networks in regulating the process of tumorigenesis and therapy response. Studies on Radiation Therapy Oncology Group tissue samples have identified aberrant expression of p53, MDM2 (an E3 ubiquitin ligase that targets p53 for proteosomal degradation), and p16 (an upstream regulator of retinoblastoma and hence E2F1 in prostate cancer); abnormal expression of these biomarkers has been associated with clinical outcome after radiotherapy ± androgen deprivation therapy. Although the proapoptotic properties of p53 are well documented, a relatively new aspect of p53 function as an active mediator of prosurvival signaling pathways is now emerging. E2F1 is a transcription factor that possesses both proapoptotic and prosurvival properties. Thus, the role of E2F1 in the process of tumorigenesis versus apoptosis is a contested issue that needs to be resolved. Furthermore, the role of E2F1 in DNA repair is being increasingly recognized. Thus, novel approaches to curb the prosurvival and DNA repair capability of E2F1 while promoting apoptotic function are of interest. In this review, we discuss the challenges involved in targeting the p53/E2F1 pathways and the crosstalk networks, and further propose potential therapeutic strategies for prostate cancer management.
Collapse
Affiliation(s)
- Thirupandiyur Udayakumar
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
10
|
Hensley HH, Hannoun-Levi JM, Hachem P, Mu Z, Stoyanova R, Khor LY, Agrawal S, Pollack A. PKA knockdown enhances cell killing in response to radiation and androgen deprivation. Int J Cancer 2010; 128:962-73. [PMID: 20960462 DOI: 10.1002/ijc.25634] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 08/03/2010] [Indexed: 01/09/2023]
Abstract
The therapeutic efficacy of Gem®231, a second generation antisense molecule targeted to the RIα subunit of PKA(RIα) (AS-PKA), administered in combination with androgen deprivation (AD) and radiation therapy (RT), was examined in androgen sensitive (LNCaP) and insensitive (PC3) cell lines. Apoptosis was assayed by Caspase 3 + 7 activity and Annexin V binding. AS-PKA significantly increased apoptosis in vitro from RT (both lines), with further increases in LNCaP cells grown in AD medium. In LNCaP cells, AD increased phosphorylated mitogen activated protein-kinase (pMAPK), which was reduced by AS-PKA relative to the mismatch (MM) controls. AS-PKA also reduced pMAPK levels in PC3 cells. Cell death was measured by clonogenic survival assays. In vivo, LNCaP cells were grown orthotopically in nude mice. Tumor kinetics were measured by magnetic resonance imaging and serum prostate-specific antigen. PC3 cells were grown subcutaneously and tumor volume assessed by caliper measurements. In PC3 xenografts, AS-PKA caused a significant increase in tumor doubling time relative to MM controls as a monotherapy or in combination with RT. In orthotopic LNCaP tumors, AS-PKA was ineffective as a monotherapy; however, it caused a statistically significant increase in tumor doubling time relative to MM controls when used in combination with AD, with or without RT. PKA(RIα) levels in tumors were quantified via immunohistochemical (IHC) staining and image analysis. IHC measurements in LNCaP cells exhibited that AS-PKA reduced PKA(RIα) levels in vivo. We demonstrate for the first time that AS-PKA enhances cell killing androgen sensitive prostate cancer cells to AD ± RT and androgen insensitive cells to RT.
Collapse
Affiliation(s)
- Harvey H Hensley
- Basic Science Division, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Signalling pathways in prostate carcinogenesis: potentials for molecular-targeted therapy. Clin Sci (Lond) 2009; 117:209-28. [PMID: 19663810 DOI: 10.1042/cs20080391] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Prostate cancer represents a major health issue and its incidence is rising globally. In developed countries, prostate cancer is the most frequently diagnosed cancer and the second most common cause of death from cancer in men. Androgen deprivation reduces tumour activity in approx. 80% of patients with advanced disease, but most tumours relapse within 2 years to an incurable hormone-resistant state. Even for patients with early disease at the time of diagnosis, a proportion of patients will unfortunately develop relapsed disease following radical therapy. Treatment options for patients with hormone-resistant prostate cancer are very limited and, even with toxic therapy, such as docetaxel, the life expectancy is only improved by a median of 2 months. Advances in molecular oncology have identified key signalling pathways that are considered to be driving events in prostate carcinogenesis. The activation of multiple signalling pathways increases further the possibility of cross-talk among 'linear' signalling cascades. Hence signalling networks that may incorporate distinct pathways in prostate cancer, particularly in hormone-resistant disease, are increasingly appreciated in drug development programmes. With the development of potent small-molecule inhibitors capable of specifically suppressing the activities of individual 'linear' cascades, it may be that, by combining these agents as guided by the molecular signature of prostate cancer, a more efficient therapeutic regime may be developed. Therefore the present review focuses on evidence of abnormal signalling in prostate cancer and the potential of these targets in drug development, and incorporates key findings of relevant clinical trials to date.
Collapse
|
12
|
Jaboin JJ, Hwang M, Perez CA, Cooper C, Chen H, Ye C, Cai Q, Wills ML, Lu B. No evidence for association of the MDM2-309 T/G promoter polymorphism with prostate cancer outcomes. Urol Oncol 2009; 29:319-23. [PMID: 19523862 DOI: 10.1016/j.urolonc.2009.03.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 03/26/2009] [Accepted: 03/31/2009] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Mouse double-minute 2 (MDM2) SNP309 polymorphism (T>G) has been correlated with an increased risk of cancer in multiple tumor types. MDM2 overexpression has shown to be weakly associated with distant tumor metastases, and down-regulation of MDM2 via antisense oligonucleotides in vitro has resulted in the radiosensitization of prostate cancer cell lines. Based on these results, we decided to evaluate the role of MDM2 SNP309 in the context of histopathologic parameters and clinical outcomes in prostate cancer tumors. MATERIALS AND METHODS The population consisted of 212 consecutive prostate cancer patients who underwent radical prostatectomy between 1997 and 1999 at Vanderbilt University Medical Center. Two hundred eight of the samples were successfully genotyped for the MDM2 SNP309 polymorphism. Correlations between the polymorphism, recurrence, and survival data were analyzed using univariate and multivariate genetic models. RESULTS The only prognostic factor predictive of overall survival in our study was Gleason score (P<0.005). Using χ(2) analysis, we determined that the MDM2 SNP309 polymorphism had no significant association with race (P=0.7512), patient's age at diagnosis (P=0.6820), pre-prostatectomy PSA level (P=0.8606), Gleason's score (P=0.4839), surgical margin status (P=1.0000), extracapsular extension (P=0 .6175), and disease stage (P=0.4945). In addition, there was no significant difference in 3-year recurrence-free survival (P=0.218), or 8-year overall survival (P=0.376). CONCLUSIONS Our study finds no evidence for association of the MDM2 SNP309 polymorphism with clinicopathologic variables, recurrence risk, and overall survival outcome in prostate cancer.
Collapse
Affiliation(s)
- Jerry J Jaboin
- Department of Radiation Oncology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Khor LY, Bae K, Paulus R, Al-Saleem T, Hammond ME, Grignon DJ, Che M, Venkatesan V, Byhardt RW, Rotman M, Hanks GE, Sandler HM, Pollack A. MDM2 and Ki-67 predict for distant metastasis and mortality in men treated with radiotherapy and androgen deprivation for prostate cancer: RTOG 92-02. J Clin Oncol 2009; 27:3177-84. [PMID: 19470936 DOI: 10.1200/jco.2008.19.8267] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE MDM2 regulates p53, which controls cell cycle arrest and apoptosis. Both proteins, along with Ki-67, which is an established strong determinant of metastasis, have shown promise in predicting the outcome of men treated with radiation therapy (RT) with or without short-term androgen deprivation (STAD). This report compares the utility of abnormal expression of these biomarkers in estimating progression in a cohort of men treated on RTOG 92-02. PATIENTS AND METHODS Adequate tissue for immunohistochemistry was available for p53, Ki-67, and MDM2 analyses in 478 patient cases. The percentage of tumor nuclei staining positive (PSP) was quantified manually or by image analysis, and the per-sample mean intensity score (MIS) was quantified by image analysis. Cox regression models were used to estimate overall mortality (OM), and Fine and Gray's regressions were applied to the end points of distant metastasis (DM) and cause-specific mortality (CSM). Results In multivariate analyses that adjusted for all markers and treatment covariates, MDM2 overexpression was significantly related to DM (P = .02) and OM (P = .003), and Ki-67 overexpression was significantly related to DM (P < .0001), CSM (P = .0007), and OM (P = .01). P53 overexpression was significantly related to OM (P = .02). When considered in combination, the overexpression of both Ki-67 and MDM2 at high levels was associated with significantly increased failure rates for all end points (P < .001 for DM, CSM, and OM). CONCLUSION Combined MDM2 and Ki-67 expression levels were independently related to distant metastasis and mortality and, if validated, could be considered for risk stratification of patients with prostate cancer in clinical trials.
Collapse
Affiliation(s)
- Li-Yan Khor
- Department of Radiation Oncology, University of Miami, Sylvester Comprehensive Cancer Center, 1475 NW 12th Ave, Suite 1501, Miami, FL 33130, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Udayakumar TS, Hachem P, Ahmed MM, Agrawal S, Pollack A. Antisense MDM2 enhances E2F1-induced apoptosis and the combination sensitizes androgen-sensitive [corrected] and androgen-insensitive [corrected] prostate cancer cells to radiation. Mol Cancer Res 2009; 6:1742-54. [PMID: 19010821 DOI: 10.1158/1541-7786.mcr-08-0102] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have previously shown in separate studies that MDM2 knockdown via antisense MDM2 (AS-MDM2) and E2F1 overexpression via adenoviral-mediated E2F1 (Ad-E2F1) sensitized prostate cancer cells to radiation. Because E2F1 and MDM2 affect apoptosis through both common and independent pathways, we hypothesized that coupling these two treatments would result in increased killing of prostate cancer cells. In this study, the effect of Ad-E2F1 and AS-MDM2 in combination with radiation was investigated in three prostate cancer cell lines: LNCaP cells, LNCaP-Res cells [androgen insensitive with functional p53 and androgen receptor (AR)], and PC3 cells (androgen insensitive, p53(null), and AR(null)). A supra-additive radiosensitizing effect was observed in terms of clonogenic inhibition and induction of apoptosis (caspase-3 + caspase-7 activity) in response to Ad-E2F1 plus AS-MDM2 treatments in all three cell lines. In LNCaP and LNCaP-Res, these combination treatments elevated the levels of phospho-Ser(15) p53 with significant induction of p21(waf1/cip1), phospho-gammaH2AX, PUMA, and Bax levels and reduction of AR and bcl-2 expression. Similarly, AR(null) and p53(null) PC-3 cells showed elevated levels of Bax and phospho-gammaH2AX expression. These findings show that the combination of Ad-E2F1 and AS-MDM2 significantly increases cell death in prostate cancer cells exposed to radiation and that this effect occurs in the presence or absence of AR and p53.
Collapse
|
15
|
Lee JT, Lehmann BD, Terrian DM, Chappell WH, Stivala F, Libra M, Martelli AM, Steelman LS, McCubrey JA. Targeting prostate cancer based on signal transduction and cell cycle pathways. Cell Cycle 2008; 7:1745-62. [PMID: 18594202 DOI: 10.4161/cc.7.12.6166] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer remains a leading cause of death in men despite increased capacity to diagnose at earlier stages. After prostate cancer has become hormone independent, which often occurs after hormonal ablation therapies, it is difficult to effectively treat. Prostate cancer may arise from mutations and dysregulation of various genes involved in regulation signal transduction (e.g., PTEN, Akt, etc.,) and the cell cycle (e.g., p53, p21(Cip1), p27(Kip1), Rb, etc.,). This review focuses on the aberrant interactions of signal transduction and cell cycle genes products and how they can contribute to prostate cancer and alter therapeutic effectiveness.
Collapse
Affiliation(s)
- John T Lee
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, North Carolina 27858, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mu Z, Hachem P, Hensley H, Stoyanova R, Kwon HW, Hanlon AL, Agrawal S, Pollack A. Antisense MDM2 enhances the response of androgen insensitive human prostate cancer cells to androgen deprivation in vitro and in vivo. Prostate 2008; 68:599-609. [PMID: 18196567 PMCID: PMC2763092 DOI: 10.1002/pros.20731] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Antisense MDM2 oligonucleotide (AS-MDM2) sensitizes androgen sensitive LNCaP cells to androgen deprivation (AD) in vitro and in vivo. In this study, we investigated the effects of AS-MDM2 combined with AD on androgen resistant LNCaP (LNCaP-Res) and moderately androgen resistant bcl-2 overexpressing LNCaP (LNCaP-BST) cells. METHODS The LNCaP-Res cell line was generated by culturing LNCaP cells in medium containing charcoal-stripped serum for more than 1 year. Apoptosis was quantified in vitro by Annexin V staining and caspase 3 + 7 activity. For the in vivo studies, orthotopic tumor growth was monitored by magnetic resonance imaging (MRI). AS-MDM2 and the mismatch control were given by i.p. injection at doses of 25 mg/kg per day, 5 days/week for 15 days. RESULTS LNCaP-Res cells expressed high levels of androgen receptor (AR) and bcl-2, and displayed no growth inhibition to AD. AS-MDM2 caused significant reductions in MDM2 and AR expression, and increases in p53 and p21 expression in both cell lines. AS-MDM2 + AD resulted in the highest levels of apoptosis in vitro and tumor growth inhibition in vivo in both cell lines; although, these effects were less pronounced in LNCaP-BST cells. CONCLUSIONS AS-MDM2 + AD enhanced apoptotic cell death in vitro and tumor growth inhibition in vivo in androgen resistant cell lines. The action of AS-MDM2 + AD was influenced somewhat by bcl-2 expression as an isolated change (LNCaP-BST cells), but not when accompanied by other molecular changes associated with androgen insensitivity (LNCaP-Res cells). MDM2 knockdown has promise for the treatment of men with early hormone refractory disease.
Collapse
Affiliation(s)
- Zhaomei Mu
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Paul Hachem
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Harvey Hensley
- Department of Basic Science, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Radka Stoyanova
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Hae Won Kwon
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Alexandra L. Hanlon
- Department of Public Education, Temple University, Philadelphia, Pennsylvania
| | | | - Alan Pollack
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Correspondence to: Alan Pollack, MD, PhD, Department of Radiation Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111.
| |
Collapse
|
17
|
Stoyanova R, Hachem P, Hensley H, Khor LY, Mu Z, Hammond MEH, Agrawal S, Pollack A. Antisense-MDM2 sensitizes LNCaP prostate cancer cells to androgen deprivation, radiation, and the combination in vivo. Int J Radiat Oncol Biol Phys 2007; 68:1151-60. [PMID: 17637390 PMCID: PMC2763094 DOI: 10.1016/j.ijrobp.2007.03.047] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 03/06/2007] [Accepted: 03/23/2007] [Indexed: 11/24/2022]
Abstract
PURPOSE To test the effects of antisense (AS)-MDM2 alone and with androgen deprivation (AD), radiotherapy (RT), and AD + RT on wild-type LNCaP cells in an orthotopic in vivo model. METHODS Androgen-sensitive LNCaP cells were grown in the prostates of nude mice. Magnetic resonance imaging-based tumor volume and serum prostate-specific antigen (PSA) measurements were used to assess effects on tumor response. Tumor response was measured by biochemical and tumor volume failure definitions and doubling time estimates from fitted PSA and tumor volume growth curves. Expression of MDM2, p53, p21, and Ki-67 was quantified using immunohistochemical staining and image analysis of formalin-fixed tissue, analogous to methods used clinically. RESULTS Antisense-MDM2 significantly inhibited the growth of LNCaP tumors over the mismatch controls. The most significant increase in tumor growth delay and tumor doubling time was from AS-MDM2 + AD + RT, although the effect of AS-MDM2 + AD was substantial. Expression of MDM2 was significantly reduced by AS-MDM2 in the setting of RT. CONCLUSIONS This is the first in vivo investigation of the effects of AS-MDM2 in an orthotopic model and the first to demonstrate incremental sensitization when added to AD and AD + RT. The results with AD underscore the potential to affect micrometastatic disease, which is probably responsible for treatment failure in 30-40% of men with high-risk disease.
Collapse
Affiliation(s)
- Radka Stoyanova
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Paul Hachem
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Harvey Hensley
- Department of Basic Science, Fox Chase Cancer Center, Philadelphia, PA
| | - Li-Yan Khor
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Zhaomei Mu
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | | | | | - Alan Pollack
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| |
Collapse
|
18
|
Lehmann BD, McCubrey JA, Terrian DM. Radiosensitization of prostate cancer by priming the wild-type p53-dependent cellular senescence pathway. Cancer Biol Ther 2007; 6:1165-70. [PMID: 18059157 DOI: 10.4161/cbt.6.8.4544] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A dramatic stage-migration in diagnosis of prostate cancer has led to earlier detection of clinically localized carcinoma and an increased use of radiation therapy. The p53 protein responds to irradiation-induced DNA damage by removing critically damaged cells from the proliferative pool. This review will focus on the dominant role that p53-dependent cellular senescence, rather than cell death, plays in determining the radiosensitivity of human prostate cancer cells in vitro. The finding that senescence is a primary mechanism of tumor regression indicates that p53 activators or downstream effectors may prove effective in radiosensitizing some carcinoma of the prostate.
Collapse
Affiliation(s)
- Brian D Lehmann
- Departments of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina, USA
| | | | | |
Collapse
|
19
|
Abstract
BACKGROUND Bcl-2 is anti-apoptotic and overexpression is associated with prostate tumor aggressiveness. We hypothesized that Bcl-2 has a role in prostate cancer radiation (RT) response. The relationship of Bcl-2 expression in four prostate cancer cell lines, and the effect of modulating expression with a Bcl-2 antisense oligonucleotide (G3139, Genasense, oblimersen sodium, Genta Incorporated), to RT was examined. METHODS The four cell lines studied were LNCaP (wild type-p53), PC3 (p53 null), Bcl-2 stably transfected LNCaP (LNCaP-BST), and Bcl-2 stably transfected PC3 (PC3-BST) cells. Cells were treated with antisense (AS) Bcl-2 alone or with RT (2-6 Gy). Following RT, cells were processed at 3-6 hr for Western blots, 18 hr for Annexin V staining and flow cytometric analysis, 24 hr for caspases 3+7 quantification by fluorometric assay, and immediately for clonogenic survival. RESULTS AS caused a significant reduction in Bcl-2 expression in all cell lines. P53 expression was elevated following RT treatment in LNCaP and LNCaP-BST cells. P21 was increased by RT treatment in all cell lines. AS caused a significant increase in caspase 3+7 activity over the mismatch (MM) controls in all cell lines. When AS was combined with RT, caspase 3+7 activity was further increased significantly over all other groups in all cell lines. Moreover, AS+RT resulted in significantly reduced clonogenic survival over MM+RT, which was dampened in the Bcl-2 overexpressing lines. CONCLUSIONS To our knowledge, these data demonstrate for the first time that a Bcl-2 specific AS oligonucleotide sensitizes prostate cancer cells to RT. p53 is not required for this effect.
Collapse
Affiliation(s)
| | | | - Alan Pollack
- Correspondence to: Alan Pollack, M.D., Ph.D., Department of Radiation Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, Phone: (215) 728-2940, Fax: (215) 728-2868, E-mail:
| |
Collapse
|
20
|
Cao C, Shinohara ET, Niermann KJ, Donnelly EF, Chen X, Hallahan DE, Lu B. Murine double minute 2 as a therapeutic target for radiation sensitization of lung cancer. Mol Cancer Ther 2005; 4:1137-45. [PMID: 16093429 DOI: 10.1158/1535-7163.mct-04-0327] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Murine double minute 2 (MDM2) inhibits p53-mediated functions, which are essential for therapies using DNA-damaging agents. The purpose of this study was to determine whether MDM2 inhibition enhances the radiosensitivity of a lung cancer model. The effects of MDM2 inhibition on tumor vasculature were also studied. Transient transfection of H460 lung cancer cells and human umbilical vascular endothelial cells (HUVEC) with antisense oligonucleotides (ASODN) against MDM2 resulted in a reduced level of MDM2 and increased levels of p21 and p53. Clonogenic assays showed that inhibition of MDM2 greatly decreased cell survival following irradiation. Quantification of apoptotic cells by 7-aminoactinomycin D staining and of senescent cells by X-gal staining showed that both processes were significantly increased in H460 cells treated with MDM2-specific ASODN and radiation. H460 xenografts that were treated with MDM2 ASODN plus radiotherapy also showed significant growth delay (P < 0.001) and increased apoptosis by terminal deoxynucleotidyl transferase-mediated nick end labeling staining. HUVECs transfected with MDM2-specific ASODN showed impaired viability and migration with decreased tube formation. Doppler studies showed that tumor blood flow was compromised when H460 xenografts were treated with MDM2-specific ASODN and radiation. A combination of radiotherapy and inhibition of MDM2 through the antisense approach results in improved tumor control in the H460 lung cancer model. This implies that a similar strategy should be investigated among patients with locally advanced lung cancer, receiving thoracic radiotherapy.
Collapse
Affiliation(s)
- Carolyn Cao
- Department of Radiation Oncology, Vanderbilt University, 1301 22nd Avenue South, B-902 The Vanderbilt Clinic, Nashville, TN 37232-5671, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Yu Y, Sun P, Sun LC, Liu GY, Chen GH, Shang LH, Wu HB, Hu J, Li Y, Mao YL, Sui GJ, Sun XW. Downregulation of MDM2 expression by RNAi inhibits LoVo human colorectal adenocarcinoma cells growth and the treatment of LoVo cells with mdm2siRNA3 enhances the sensitivity to cisplatin. Biochem Biophys Res Commun 2005; 339:71-8. [PMID: 16297858 DOI: 10.1016/j.bbrc.2005.10.181] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2005] [Accepted: 10/21/2005] [Indexed: 10/25/2022]
Abstract
To investigate the biological effect of mdm2 in human colorectal adenocarcinoma LoVo cells, three mdm2siRNA constructions were recombined and transient transfected into human colorectal adenocarcinoma LoVo cells with low differentiation character in vitro. The results showed that mdm2siRNA3 reduced mRNA level of mdm2 and protein level of mdm2, leading to proliferation inhibition on LoVo cells, and reduced tumor growth in nude mice. It was found that depletion of MDM2 in this pattern promoted apoptosis of LoVo cells and Cisplatin (DDP) treated in the mdm2siRNA3 transfected cell population would result in a substantial decrease by MTT colorimetry. Decreasing the MDM2 protein level in LoVo cells by RNAi could significantly inhibit tumor growth both in vitro and in vivo, which indicated that mdm2 gene played a definite role in the development and aggressiveness of human colon carcinoma. It also could be a therapeutic target in colorectal carcinoma. The synergistic activation of RNAi and cell toxicity agents indicated that the combination of chemotherapy and gene therapy will be a promising approach in the future.
Collapse
Affiliation(s)
- Yan Yu
- Department of Internal Medicine, Cancer Hospital of Harbin Medical University, Harbin 150040, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Nguyen KH, Hachem P, Khor LY, Salem N, Hunt KK, Calkins PR, Pollack A. Adenoviral-E2F-1 radiosensitizes p53wild-type and p53null human prostate cancer cells. Int J Radiat Oncol Biol Phys 2005; 63:238-46. [PMID: 15993550 PMCID: PMC4347813 DOI: 10.1016/j.ijrobp.2005.04.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 04/22/2005] [Accepted: 04/26/2005] [Indexed: 01/30/2023]
Abstract
PURPOSE E2F-1 is a transcription factor that enhances the radiosensitivity of various cell lines by inducing apoptosis. However, there are conflicting data concerning whether this enhancement is mediated via p53 dependent pathways. Additionally, the role of E2F-1 in the response of human prostate cancer to radiation has not been well characterized. In this study, we investigated the effect of Adenoviral-E2F-1 (Ad-E2F-1) on the radiosensitivity of p53wild-type (LNCaP) and p53null (PC3) prostate cancer cell lines. METHODS AND MATERIALS LNCaP and PC3 cells were transduced with Ad-E2F-1, Adenoviral-Luciferase (Ad-Luc) control vector, or Adenoviral-p53 (Ad-p53). Expression of E2F-1 and p53 was examined by Western blot analysis. Annexin V and caspase 3 + 7 assays were performed to estimate the levels of apoptosis. Clonogenic survival assays were used to determine overall cell death. Statistical significance was determined by analysis of variance, using the Bonferroni method to correct for multiple comparisons. RESULTS Western blot analysis confirmed the efficacy of transductions with Ad-E2F-1 and Ad-p53. Ad-E2F-1 transduction significantly enhanced apoptosis and decreased clonogenic survival in both cell lines. These effects were compounded by the addition of RT. Although E2F-1-mediated radiosensitization was independent of p53 status, this effect was more pronounced in p53wild-type LNCaP cells. When PC3 cells were treated with Ad-p53 in combination with RT and Ad-E2F-1, there was at least an additive reduction in clonogenic survival. CONCLUSIONS Our results suggest that Ad-E2F-1 significantly enhances the response of p53wild-type and p53null prostate cancer cells to radiation therapy, although radiosensitization is more pronounced in the presence of p53. Ad-E2F-1 may be a useful adjunct to radiation therapy in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Khanh H. Nguyen
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Paul Hachem
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Li-Yan Khor
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Naji Salem
- Department of Radiotherapy, Institut Paoli-Calmette, Avignon, France
| | - Kelly K. Hunt
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Peter R. Calkins
- Department of Pathology, Baylor College of Medicine, Houston, TX
| | - Alan Pollack
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA
| |
Collapse
|
23
|
Freytag SO, Kim JH, Brown SL, Barton K, Lu M, Chung M. Gene therapy strategies to improve the effectiveness of cancer radiotherapy. Expert Opin Biol Ther 2005; 4:1757-70. [PMID: 15500404 DOI: 10.1517/14712598.4.11.1757] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Having the ability to alter the genetic makeup of a cancer cell by gene transfer is a potentially powerful strategy for treating human cancer. However, a low efficiency of gene delivery in vivo and poor tumour specificity has prevented the widespread implementation of this technology in the clinic. Despite these formidable obstacles, the first successful application of gene therapy in the treatment of cancer may occur when it is combined with local modalities such as radiation therapy. A small number of gene therapy strategies have been evaluated in clinical trials in combination with external beam radiation therapy. The combined therapy has been well-tolerated and has not exacerbated the side effects of radiation therapy. Gene transfer and tumour cell destruction has been demonstrated in vivo. Although the results await confirmation in larger, prospective Phase III trials, there is suggestive evidence that the combined therapies may be demonstrating better than expected antitumour activity. Our vast knowledge of the molecular defects that drive the cancer process, coupled with our expanding understanding of the genes responsible for tumour cell radioresistance, have spawned the development of rational, targeted gene therapies designed to increase tumour cell radiosensitivity. Here, the results of the clinical trials conducted so far will be reviewed, followed by a description of new approaches under development at present.
Collapse
Affiliation(s)
- Svend O Freytag
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI 48202-3405, USA.
| | | | | | | | | | | |
Collapse
|
24
|
McCarty MF. Targeting multiple signaling pathways as a strategy for managing prostate cancer: multifocal signal modulation therapy. Integr Cancer Ther 2005; 3:349-80. [PMID: 15523106 DOI: 10.1177/1534735404270757] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The aberrant behavior of cancer reflects upregulation of certain oncogenic signaling pathways that promote proliferation, inhibit apoptosis, and enable the cancer to spread and evoke angiogenesis. Theoretically, it should be feasible to decrease the activity of these pathways-or increase the activity of pathways that oppose them-with noncytotoxic agents. Since multiple pathways are dysfunctional in most cancers, and cancers accumulate new oncogenic mutations as they progress, the greatest and most durable therapeutic benefit will likely be achieved with combination regimens that address several targets. Thus, a multifocal signal modulation therapy (MSMT) of cancer is proposed. This concept has already been documented by researchers who have shown that certain combinations of signal modulators-of limited utility when administered individually-can achieve dramatic suppression of tumor growth in rodent xenograft models. The present essay attempts to guide development of MSMTs for prostate cancer. Androgen ablation is a signal-modulating measure already in standard use in the management of delocalized prostate cancer. The additional molecular targets considered here include the type 1 insulin-like growth factor receptor, the epidermal growth factor receptor, mammalian target of rapamycin, NF-kappaB, hypoxia-inducible factor-1alpha, hsp90, cyclooxygenase-2, protein kinase A type I, vascular endothelial growth factor, 5-lipoxygenase, 12-lipoxygenase, angiotensin II receptor type 1, bradykinin receptor type 1, c-Src, interleukin-6, ras, MDM2, bcl-2/bclxL, vitamin D receptor, estrogen receptor-beta, and PPAR-. Various nutrients and phytochemicals suspected to have potential utility in prostate cancer prevention and therapy, but whose key molecular targets are still unknown, might reasonably be incorporated into MSMTs for prostate cancer; these include lycopene, selenium, green tea polyphenols, genistein, and silibinin. MSMTs can be developed systematically by testing various combinations of signal-modulating agents, in concentrations that can feasibly be achieved and maintained clinically, on human prostate cancer cell lines; combinations that appear promising can then be tested in xenograft models and, ultimately, in the clinic. Some signal modulators can increase response to cytotoxic drugs by upregulating effectors of apoptosis. When MSMTs fail to raise the spontaneous apoptosis rate sufficiently to achieve tumor stasis or regression, incorporation of appropriate cytotoxic agents into the regimen may improve the clinical outcome.
Collapse
|
25
|
Khor LY, Desilvio M, Al-Saleem T, Hammond ME, Grignon DJ, Sause W, Pilepich M, Okunieff P, Sandler H, Pollack A. MDM2 as a predictor of prostate carcinoma outcome. Cancer 2005; 104:962-7. [PMID: 16007688 DOI: 10.1002/cncr.21261] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND The MDM2 oncoprotein promotes p53 degradation via ubiquitin, establishing negative feedback control of p53 and consequently affecting cell cycle arrest and apoptosis. The authors evaluated the association between MDM2 expression and local failure, distant metastasis (DM), cause-specific mortality, and overall mortality in men treated in Radiation Therapy Oncology Group 8610 with radiotherapy, with or without androgen deprivation. METHODS Of the 456 eligible and analyzable patients (parent cohort), adequate archival diagnostic tissue specimens from 108 patients were available for MDM2 analysis (MDM2 cohort). Cox proportional hazards multivariate analysis (MVA) was used to determine the relation of MDM2 to the endpoints. MDM2 overexpression was manually classified as > 5% nuclear staining. An image analysis system was also used to quantify the proportion of tumor nuclei with MDM2 staining (ACIS index) and staining intensity. RESULTS Overexpression of MDM2 by manual counts was seen in 44% (n = 47) of the patients. In the manual count analysis, there was no significant relation between MDM2 overexpression and outcome. The ACIS index, using a cutoff point defined by the median value, < or = 3% versus > 3%, was related to 5-year DM rates in univariate analyses (32.6% vs. 45.8%; P = 0.057) and MVA (P = 0.06). The intensity of MDM2 staining was not significant. CONCLUSIONS MDM2 expression quantified by image analysis was weakly associated with DM. The cohort examined was relatively small and with larger patient numbers, MDM2 overexpression may emerge as a more significant covariate.
Collapse
Affiliation(s)
- Li-Yan Khor
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|