1
|
Freeman ML. Oncology Scan—The Concept of Personalized Medicine and the Radiation Response of Tumors. Int J Radiat Oncol Biol Phys 2014; 88:546-8. [DOI: 10.1016/j.ijrobp.2013.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Indexed: 10/25/2022]
|
2
|
Kijima T, Koga F, Fujii Y, Yoshida S, Tatokoro M, Kihara K. Zoledronic acid sensitizes renal cell carcinoma cells to radiation by downregulating STAT1. PLoS One 2013; 8:e64615. [PMID: 23741352 PMCID: PMC3669422 DOI: 10.1371/journal.pone.0064615] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 04/15/2013] [Indexed: 11/25/2022] Open
Abstract
Zoledronic acid (ZOL), a third-generation bisphosphonate that strongly inhibits osteoclast activity, is widely used for the treatment of bone metastasis from a variety of malignancies, including renal cell carcinoma (RCC). We previously reported that zoledronic acid (ZOL) clinically potentiates antitumor effects of radiotherapy (RT) on bone metastases from RCC. To date, however, it remains unknown whether ZOL radiosensitizes RCC and if it does, how. Here, we demonstrated that ZOL directly radiosensitizes RCC cells independent of osteoclast activity by potentiating the caspase-3-mediated apoptosis pathway. The radiosensitization by ZOL was observed in 786-O, A-498, and ACHN cells but not in Caki-1 cells. As its underlying molecular mechanism, we found that the signal transducer and activator of transcription 1 (STAT1) plays a key role. The three RCC cell lines, in which ZOL exerted a radiosensitizing effect, expressed STAT1 abundantly but Caki-1 cells did not. ZOL downregulated endogenous STAT1 expression in 786-O, A-498, and ACHN cells by a post-transcriptional modification. We confirmed that knockdown of endogenous STAT1 by siRNA sensitized 786-O cells to RT equivalently to ZOL, and that introduction of exogenous STAT1 rendered Caki-1 cells more RT-resistant. This is the first study to clarify the molecular mechanism by which ZOL directly radiosensitizes tumor cells. Because tumor cells commonly overexpress STAT1 and ZOL reportedly radiosensitizes various types of tumor cells, ZOL warrants further clinical and translational studies as a potent radiosensitizer against RT-resistant tumors overexpressing STAT1.
Collapse
Affiliation(s)
- Toshiki Kijima
- Graduate School of the Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fumitaka Koga
- Graduate School of the Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| | - Yasuhisa Fujii
- Graduate School of the Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Soichiro Yoshida
- Graduate School of the Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Manabu Tatokoro
- Graduate School of the Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazunori Kihara
- Graduate School of the Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
3
|
Langlands FE, Horgan K, Dodwell DD, Smith L. Breast cancer subtypes: response to radiotherapy and potential radiosensitisation. Br J Radiol 2013; 86:20120601. [PMID: 23392193 DOI: 10.1259/bjr.20120601] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Radiotherapy (RT) is of critical importance in the locoregional management of early breast cancer. Over 50% of patients receive RT at some time during the treatment of their disease, equating to over 500 000 patients worldwide receiving RT each year. Unfortunately, not all patients derive therapeutic benefit and some breast cancers are resistant to treatment, as evidenced by distant metastatic spread and local recurrence. Prediction of individual responses to RT may allow a stratified approach to this treatment permitting those patients with radioresistant tumours to receive higher doses of RT (total and/or tumour cavity boost doses) and/or radiosensitising agents to optimise treatment. Also, for those patients unlikely to respond at all, it would prevent harmful side effects occurring for no therapeutic gain. More selective targeting would better direct National Health Service resources, ease the burden on heavily used treatment RT machines and reduce the economic cost of cancer treatment. Unfortunately, there are no robust and validated biomarkers for predicting RT outcome. We review the available literature to determine whether classification of breast cancers according to their molecular profile may be used to predict successful response to, or increased morbidity from, RT. Class-specific biomarkers for targeting by radiosensitising agents are also discussed.
Collapse
Affiliation(s)
- F E Langlands
- Section of Pathology and Tumour Biology, Leeds Institute of Molecular Medicine, Leeds University, Leeds, UK
| | | | | | | |
Collapse
|
4
|
Karroum O, Kengen J, Danhier P, Magat J, Mignion L, Bouzin C, Verrax J, Charette N, Starkel P, Calderon PB, Sonveaux P, Feron O, Grégoire V, Gallez B, Jordan BF. Tumor reoxygenation following administration of Mitogen-Activated Protein Kinase inhibitors: a rationale for combination with radiation therapy. Radiother Oncol 2012; 105:64-71. [PMID: 22682746 DOI: 10.1016/j.radonc.2012.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 04/12/2012] [Accepted: 05/14/2012] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE The relevance of Mitogen Activated Protein Kinase (MAPK) inhibitors as co-treatments for radiation therapy is investigated, with special focus on a potential link between the MAPK pathway and tumor hypoxia, which is a critical determinant for response to therapy. MATERIALS AND METHODS The effects of two MAPK inhibitors, Sorafenib and PD0325901, were monitored daily using in vivo EPR (Electron Paramagnetic Resonance) oximetry in FSaII and TLT tumor models in order to identify a window of reoxygenation, during which tumor blood flow, oxygen consumption and radiation sensitivity were assessed. RESULTS Reoxygenation was shown after two days of treatments with Sorafenib or PD0325901 in two tumor models, which was further successfully exploited with Sorafenib for improving the radiation response of FSaII tumors by a factor of 1.5. The increase in tumor oxygenation was shown to be the result of two major factors: (i) an increase in blood flow for Sorafenib, that might be linked to its anti-angiogenic effect (vascular normalization), and (ii) a decrease in oxygen consumption for Sorafenib and PD0325901, due to an alteration of the mitochondrial activity. CONCLUSION We evidenced tumor reoxygenation in vivo following MAPK inhibition and suggest a rationale for the combination of radiation therapy with Sorafenib.
Collapse
|
5
|
Krens LL, Baas JM, Gelderblom H, Guchelaar HJ. Therapeutic modulation of k-ras signaling in colorectal cancer. Drug Discov Today 2010; 15:502-16. [PMID: 20594936 DOI: 10.1016/j.drudis.2010.05.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 04/14/2010] [Accepted: 05/21/2010] [Indexed: 02/07/2023]
Abstract
KRAS has an important role in colorectal carcinogenesis and mutant KRAS leads to a permanently activated k-ras protein. To exert its biological activity, k-ras requires post-translational modification by prenylation. K-ras modulation has become a promising concept for new therapies, mostly by interference with the mevalonate pathway and subsequently by the prenylation of k-ras. Clinical data of agents interfering with the mevalonate pathway and the prenylation of ras are summarized and suggest that these agents might be effective when administered in combination with anticancer drugs that target k-ras. Here, we discuss the novel concept that modulation of k-ras might potentiate EGFR therapy by altering the KRAS phenotype.
Collapse
Affiliation(s)
- Lisanne L Krens
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|
6
|
Yu M, Han J, Dai M, Cui P, Li H, Liu Q, Xiu R. Influence of PEG-conjugated hemoglobin on tumor oxygenation and response to chemotherapy. ACTA ACUST UNITED AC 2008; 36:551-61. [PMID: 19065307 DOI: 10.1080/10731190802556674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Hypoxic tumors are significantly more malignant, metastatic, radio- and chemoresistant. The use of artificial oxygen carriers represents a new approach to the problem of hypoxia. In the present study, female athymic BALB/c nude mice bearing the cervical carcinoma were untreated or treated with cisplatin to determine whether administration of artificial oxygen carrier (PEG-conjugated Hemoglobin, PEG-Hb) could improve the tumor oxygenation and enhance the anti-tumor efficacy of cisplatin. Pimonidazole staining was employed to detect tumor tissue oxygenation status. We found that the application of a higher dose (0.6 g/kg) PEG-Hb could significantly ameliorate the hypoxic condition in cervical carcinoma xenograft models. Co-administration of PEG-Hb (0.6 g/kg) with cisplatin produced significant tumor growth inhibition and pro-apoptotic and anti-proliferative effects as compared to cisplatin alone. These suggest the evaluated PEG-Hb in this experiment has positive effects on cisplatin or cisplatin-based chemotherapy, and further work to optimize its application is warranted.
Collapse
Affiliation(s)
- Minghua Yu
- Institute of Microcirculation, Peking Union Medical College & Chinese Academy of Medical Sciences, 5 Dong Dan San Tiao, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
7
|
Torres-Roca JF, Stevens CW. Predicting response to clinical radiotherapy: past, present, and future directions. Cancer Control 2008; 15:151-6. [PMID: 18376382 DOI: 10.1177/107327480801500207] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Personalized radiation therapy holds the promise that the diagnosis, prevention, and treatment of cancer will be based on individual assessment of risk. Although advances in personalized radiation therapy have been achieved, the biological parameters that define individual radiosensitivity remain unclear. METHODS This review focuses on discussing the field of radiosensitivity predictive assays, a technology central to the concept of personalized medicine in radiation oncology. Two novel approaches, DNA end-binding complexes and gene expression classifiers, show promise in solving some of the logistic problems associated with previous assays. RESULTS Current data suggest that predicting clinical response to radiotherapy is possible. The delivery of this promise depends on the ability to define the variables that define response to clinical radiotherapy. A successful predictive assay is key to the development of personalized treatment strategies in radiation oncology. CONCLUSIONS Novel technologies need to be developed that will improve our understanding of the biological variables that define clinical tumor response and will lead to the development of a clinically useful assay.
Collapse
Affiliation(s)
- Javier F Torres-Roca
- Radiation Oncology Program, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | | |
Collapse
|
8
|
Effect of proton beam on blood vessel formation in early developing zebrafish (Danio rerio) embryos. Arch Pharm Res 2008; 31:779-85. [PMID: 18563361 DOI: 10.1007/s12272-001-1226-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 04/13/2008] [Accepted: 05/08/2008] [Indexed: 10/21/2022]
Abstract
Proton beam therapy can kill tumor cells while saving normal cells because of its specific energy delivery properties and so is used to various tumor patients. However, the effect of proton beam on angiogenesis in the development of blood vessels has not been determined. Here we used the zebrafish model to determine in vivo whether proton beam inhibits angiogenesis. Flk-1-GFP transgenic embryos irradiated with protons (35 MeV, spread out Bragg peak, SOBP) demonstrated a marked inhibition of embryonic growth and an altered fluorescent blood vessel development in the trunk region. When cells were stained with acridine orange to evaluate DNA damage, the number of green fluorescent cell death spots was increased in trunk regions of irradiated embryos compared to non-irradiated control embryos. Proton beam also significantly increased the cell death rate in human umbilical vein endothelial cells (HUVEC), but pretreatment with N-acetyl cystein (NAC), an antioxidant, reduced the proton-induced cell death rate (p<0.01). Moreover, pretreatment with NAC abrogated the inhibition of trunk vessel development and prevented the trunk malformation caused by proton irradiation. In conclusion, proton irradiation significantly inhibited in vivo vascular development possibly due to increased vascular cell death via reactive oxygen species formation.
Collapse
|
9
|
Choi S, Myers JN. Molecular pathogenesis of oral squamous cell carcinoma: implications for therapy. J Dent Res 2008; 87:14-32. [PMID: 18096889 DOI: 10.1177/154405910808700104] [Citation(s) in RCA: 323] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The development of oral squamous cell carcinoma (OSCC) is a multistep process requiring the accumulation of multiple genetic alterations, influenced by a patient's genetic predisposition as well as by environmental influences, including tobacco, alcohol, chronic inflammation, and viral infection. Tumorigenic genetic alterations consist of two major types: tumor suppressor genes, which promote tumor development when inactivated; and oncogenes, which promote tumor development when activated. Tumor suppressor genes can be inactivated through genetic events such as mutation, loss of heterozygosity, or deletion, or by epigenetic modifications such as DNA methylation or chromatin remodeling. Oncogenes can be activated through overexpression due to gene amplification, increased transcription, or changes in structure due to mutations that lead to increased transforming activity. This review focuses on the molecular mechanisms of oral carcinogenesis and the use of biologic therapy to specifically target molecules altered in OSCC. The rapid progress that has been made in our understanding of the molecular alterations contributing to the development of OSCC is leading to improvements in the early diagnosis of tumors and the refinement of biologic treatments individualized to the specific characteristics of a patient's tumor.
Collapse
Affiliation(s)
- S Choi
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 441, [corrected] Houston, TX 77030-4009, USA
| | | |
Collapse
|
10
|
Growth inhibition of non-small-cell lung carcinoma by BN/GRP antagonist is linked with suppression of K-Ras, COX-2, and pAkt. Proc Natl Acad Sci U S A 2007; 104:18671-6. [PMID: 18003891 DOI: 10.1073/pnas.0709455104] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bombesin (BN) or gastrin-releasing peptide (GRP) can stimulate the growth of neoplasms such as breast cancer and small-cell lung carcinoma (SCLC). Antagonists of BN/GRP have been shown to inhibit these cancers. We evaluated whether antagonists of BN/GRP can suppress the growth of human non-SCLC (NSCLC) xenografted into nude mice. The effect of the administration of BN/GRP antagonist RC-3940-II on the growth of H460 and A549 NSCLC cell lines orthotopically xenografted into the intrapulmonary interstitium was examined. Protein levels of K-Ras, COX-2, Akt/pAkt, WT p53, Erk1/2, and lung resistance-related protein (LRP) in tumors were analyzed by Western blot analaysis, and receptors for BN/GRP were investigated by radioligand-binding studies. The effect of RC-3940-II on the proliferation of H460 and A549 cells in vitro was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assays. High-affinity receptors for BN/GRP were found on tumors. Treatment with RC-3940-II significantly (P < 0.001) inhibited growth of H460 and A549 NSCLC xenografts by 30-50% and led to an improved performance status, compared with controls. In H460 NSCLC, the antitumor effect was associated with a significant (P < 0.001) reduction in protein levels of K-Ras, COX-2, pAkt, and pERK1/2 and with a major augmentation in the expression of WT p53, compared with controls. In A549 NSCLC, pAkt and LRP were significantly down-regulated. Our findings demonstrate the efficacy of BN/GRP antagonist RC-3940-II for the treatment of NSCLC. The suppression of K-Ras, COX-2, pAkt, and LRP, as well as the up-regulation of WT p53 might contribute to the antitumor action of BN/GRP antagonists.
Collapse
|
11
|
Konstantinopoulos PA, Karamouzis MV, Papavassiliou AG. Post-translational modifications and regulation of the RAS superfamily of GTPases as anticancer targets. Nat Rev Drug Discov 2007; 6:541-55. [PMID: 17585331 DOI: 10.1038/nrd2221] [Citation(s) in RCA: 355] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The involvement of the RAS superfamily of monomeric GTPases in carcinogenesis is increasingly being appreciated. A complex array of post-translational modifications and a highly sophisticated protein network regulate the spatio-temporal activation of these GTPases. Previous attempts to pharmacologically target this family have focused on the development of farnesyltransferase inhibitors, but the performance of such agents in cancer clinical trials has not been as good as hoped. Here, we review emerging druggable targets and novel therapeutic approaches targeting prenylation and post-prenylation modifications and the functional regulation of GDP/GTP exchange as exciting alternatives for anticancer therapy.
Collapse
|
12
|
Bristow RG, Ozcelik H, Jalali F, Chan N, Vesprini D. Homologous recombination and prostate cancer: a model for novel DNA repair targets and therapies. Radiother Oncol 2007; 83:220-30. [PMID: 17531338 DOI: 10.1016/j.radonc.2007.04.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2007] [Accepted: 04/17/2007] [Indexed: 01/30/2023]
Abstract
Using elegant targeting techniques such as IMRT, radiation oncology has improved the therapeutic ratio of prostate cancer radiotherapy through increased physical precision (e.g. increased local control through dose-escalation without increased normal tissue toxicity). The therapeutic ratio might be further improved by the addition of "biologic precision and escalation" pertaining to the use of molecular inhibitors of DNA damage sensing and repair. Indeed, proteins involved in the ATM-p53 damage signaling axis and the homologous (HR) and non-homologous end-joining (NHEJ) pathways of DNA double-strand break (DNA-dsb) rejoining pathways may be attractive candidates to elucidate cancer risk, prognosis, prediction of response and to develop sensitizers towards oxic and hypoxic prostate tumor cells. This review highlights DNA-dsb in prostate cancer research in terms of novel molecular inhibitors, the role of the microenvironment in DNA-dsb repair and potential DNA-dsb biomarkers for clinical trials.
Collapse
Affiliation(s)
- Robert G Bristow
- Department of Medical Biophysics, University of Toronto and Princess Margaret Hospital (University Health Network), Toronto, Canada.
| | | | | | | | | |
Collapse
|
13
|
Bentzen SM, Harari PM, Bernier J. Exploitable mechanisms for combining drugs with radiation: concepts, achievements and future directions. ACTA ACUST UNITED AC 2007; 4:172-80. [PMID: 17327857 DOI: 10.1038/ncponc0744] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Accepted: 10/06/2006] [Indexed: 12/27/2022]
Abstract
Widening indications for combining radiation therapy with cytotoxic or molecular-targeted drugs have mainly been driven by pragmatic clinical trials. With a flurry of novel drugs in various stages of preclinical and clinical development there is a need to revise the framework that has traditionally been used for discussing possible drug-radiation interactions, especially because many of the new drugs are directed at a specific molecular target. Spatial cooperation, cytotoxic enhancement, biological cooperation, temporal modulation and normal tissue protection are proposed as five primary exploitable mechanisms for the rational combination of drugs with radiation for cancer therapy. These five mechanisms produce different clinical outcomes and, therefore, the optimum clinical end point for assessing therapeutic benefit will depend on the mechanism tested. The dependence of outcome on these mechanisms also affects the selection of preclinical models and the optimum scheduling of the two modalities, i.e. the timing and dosing of the drug relative to the radiation dose fractions. These considerations are discussed in some detail for each mechanism and illustrated with specific clinical examples. Multi-modality therapy for head and neck squamous-cell carcinoma is used to illustrate these concepts. Further clinical progress in this field will require hypothesis-driven trials to ensure efficient identification of treatments with the most favorable risk:benefit ratio.
Collapse
Affiliation(s)
- Søren M Bentzen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, K4/316 Clinical Sciences Center, Madison, WI 53792, USA.
| | | | | |
Collapse
|
14
|
Hohla F, Schally AV, Szepeshazi K, Varga JL, Buchholz S, Köster F, Heinrich E, Halmos G, Rick FG, Kannadka C, Datz C, Kanashiro CA. Synergistic inhibition of growth of lung carcinomas by antagonists of growth hormone-releasing hormone in combination with docetaxel. Proc Natl Acad Sci U S A 2006; 103:14513-8. [PMID: 16983095 PMCID: PMC1599991 DOI: 10.1073/pnas.0605309103] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We investigated the effect of antagonists of growth hormone-releasing hormone (GHRH) MZ-J-7-138 and JV-1-92 on H460 human non-small cell lung carcinoma (NSCLC) xenografted orthotopically into nude mice. Treatment with MZ-J-7-138 or JV-1-92 inhibited orthotopic growth of H460 NSCLC by 52-65% (P < 0.001) and was associated with a significant decrease in protein expression of K-Ras, cyclooxygenase-2 (Cox-2) and phospho-Akt (pAkt). In other experiments, treatment with MZ-J-7-138 or docetaxel reduced tumor volume of s.c. xenografted H460 human NSCLC by 30-36% (P < 0.01). The combination of MZ-J-7-138 and docetaxel resulted in a synergistic growth inhibition of H460 NSCLC xenografts of 63%. MZ-J-7-138 alone or in combination with docetaxel significantly reduced protein levels of K-Ras, Cox-2, and pAkt by 56-63%. Docetaxel given singly diminished the protein levels only of Cox-2 and did not affect K-Ras and pAkt. High-affinity binding sites, mRNA, and protein expression of pituitary GHRH receptors and its splice variant (SV) 1 were found in H460. H460 NSCLC cells contained GHRH peptide, and its growth was significantly inhibited in vitro by 10 microM MZ-J-7-138 (P < 0.001). Serum insulin-like growth factor 1 (IGF1) was not reduced by either GHRH antagonists. These findings suggest that antiproliferative effects of GHRH antagonists in H460 NSCLC are associated with down-regulation of K-Ras, Cox-2, and pAkt. In conclusion, GHRH antagonists in combination with docetaxel synergistically inhibit growth of H460 NSCLC and the expression of K-ras, Cox-2, and pAkt, which might abrogate the signal transduction pathways for cell growth stimulation and therapeutic resistance.
Collapse
MESH Headings
- Alternative Splicing/genetics
- Animals
- Antineoplastic Agents, Phytogenic/therapeutic use
- Body Weight/drug effects
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Cell Proliferation/drug effects
- Cyclooxygenase 2/metabolism
- Docetaxel
- Drug Synergism
- Gene Expression Regulation, Neoplastic/drug effects
- Growth Hormone-Releasing Hormone/antagonists & inhibitors
- Humans
- Insulin-Like Growth Factor I/metabolism
- Mice
- Mice, Nude
- Organ Size/drug effects
- Phosphoproteins/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins p21(ras)/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Radioimmunoassay
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/genetics
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- Taxoids/therapeutic use
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Florian Hohla
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- Department of Internal Medicine, Hospital Oberndorf, 5100 Oberndorf, Austria
| | - Andrew V. Schally
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125
- University of Miami Miller School of Medicine, Miami, FL 33101; and
| | - Karoly Szepeshazi
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125
| | - Jozsef L. Varga
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Stefan Buchholz
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Frank Köster
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Elmar Heinrich
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Gabor Halmos
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- University of Miami Miller School of Medicine, Miami, FL 33101; and
| | - Ferenc G. Rick
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Chandrika Kannadka
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Christian Datz
- Department of Internal Medicine, Hospital Oberndorf, 5100 Oberndorf, Austria
| | - Celia A. Kanashiro
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| |
Collapse
|
15
|
Cairns R, Papandreou I, Denko N. Overcoming physiologic barriers to cancer treatment by molecularly targeting the tumor microenvironment. Mol Cancer Res 2006; 4:61-70. [PMID: 16513837 DOI: 10.1158/1541-7786.mcr-06-0002] [Citation(s) in RCA: 260] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is widely recognized that the vasculature of the tumor is inadequate to meet the demands of the growing mass. The malformed vasculature is at least in part responsible for regions of the tumor that are hypoxic, acidotic, and exposed to increased interstitial fluid pressure. These unique aspects of the tumor microenvironment have been shown to act as barriers to conventional chemotherapy or radiation-based therapies. It now seems that while the vasculature initiates these tumor-specific conditions, the cells within the tumor respond to these stresses and add to the unique solid tumor physiology. Gene expression changes have been reported in the tumor for vascular endothelial growth factor, carbonic anhydrase IX, and pyruvate dehydrogenase kinase 1. The activity of these gene products then influences the tumor physiology through alterations in vascular permeability and interstitial fluid pressure, extracellular acidosis, and mitochondrial oxygen consumption and hypoxia, respectively. Novel molecular strategies designed to interfere with the activities of these gene products are being devised as ways to overcome the physiologic barriers in the tumor to standard anticancer therapies.
Collapse
Affiliation(s)
- Rob Cairns
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, CCSR-South, Room 1245, Stanford, CA 94305-5152, USA
| | | | | |
Collapse
|
16
|
Basso AD, Kirschmeier P, Bishop WR. Thematic review series: Lipid Posttranslational Modifications. Farnesyl transferase inhibitors. J Lipid Res 2006; 47:15-31. [PMID: 16278491 DOI: 10.1194/jlr.r500012-jlr200] [Citation(s) in RCA: 232] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Some proteins undergo posttranslational modification by the addition of an isoprenyl lipid (farnesyl- or geranylgeranyl-isoprenoid) to a cysteine residue proximal to the C terminus. Protein isoprenylation promotes membrane association and contributes to protein-protein interactions. Farnesylated proteins include small GTPases, tyrosine phosphatases, nuclear lamina, cochaperones, and centromere-associated proteins. Prenylation is required for the transforming activity of Ras. Because of the high frequency of Ras mutations in cancer, farnesyl transferase inhibitors (FTIs) were investigated as a means to antagonize Ras function. Evaluation of FTIs led to the finding that both K- and N-Ras are alternatively modified by geranylgeranyl prenyltransferase-1 in FTI-treated cells. Geranylgeranylated forms of Ras retain the ability to associate with the plasma membrane and activate substrates. Despite this, FTIs are effective at inhibiting the growth of human tumor cells in vitro, suggesting that activity is dependent on blocking the farnesylation of other proteins. FTIs also inhibit the in vivo growth of human tumor xenografts and sensitize these models to chemotherapeutics, most notably taxanes. Several FTIs have entered clinical trials for various cancer indications. In some clinical settings, primarily hematologic malignancies, FTIs have displayed evidence of single-agent activity. Clinical studies in progress are exploring the antitumor activity of FTIs as single agents and in combination. This review will summarize the basic biology of FTIs, their antitumor activity in preclinical models, and the current status of clinical studies with these agents.
Collapse
Affiliation(s)
- Andrea D Basso
- Schering-Plough Research Institute, Kenilworth, NJ 07033, USA.
| | | | | |
Collapse
|
17
|
Kim IA, Bae SS, Fernandes A, Wu J, Muschel RJ, McKenna WG, Birnbaum MJ, Bernhard EJ. Selective inhibition of Ras, phosphoinositide 3 kinase, and Akt isoforms increases the radiosensitivity of human carcinoma cell lines. Cancer Res 2005; 65:7902-10. [PMID: 16140961 DOI: 10.1158/0008-5472.can-05-0513] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ras activation promotes the survival of tumor cells after DNA damage. To reverse this survival advantage, Ras signaling has been targeted for inhibition. Other contributors to Ras-mediated DNA damage survival have been identified using pharmacologic inhibition of signaling, but this approach is limited by the specificity of the inhibitors used and their toxicity. To better define components of Ras signaling that could be inhibited in a clinical setting, RNA interference was used to selectively block expression of specific isoforms of Ras, phosphoinositide 3 (PI3) kinase, and Akt. Inhibition of oncogenic Ras expression decreased both phospho-Akt and phospho-p42/44 mitogen-activated protein (MAP) kinase levels and reduced clonogenic survival. Because pharmacologic inhibition of PI3 kinases and Akt radiosensitized cell lines with active Ras signaling, whereas inhibition of the MAP/extracellular signal-regulated kinase (ERK) kinase/ERK pathway did not, we examined the contribution of PI3 kinases and Akts to radiation survival. Selective inhibition the PI3 kinase P110alpha + p85beta isoforms reduced Akt phosphorylation and radiation survival. Similarly, inhibition of Akt-1 reduced tumor cell radiation survival. Inhibition of Akt-2 or Akt-3 had less effect. Retroviral transduction and overexpression of mouse Akt-1 was shown to rescue cells from inhibition of endogenous human Akt-1 expression. This study shows that Ras signaling to the PI3 kinase-Akt pathway is an important contributor to survival, whether Ras activation results from mutation of ras or overexpression of epidermal growth factor receptor. This study further shows that selective inhibition of the PI3 kinase P110alpha + p85beta isoforms or Akt-1 could be a viable approach to sensitizing many tumor cells to cytotoxic therapies.
Collapse
Affiliation(s)
- In-Ah Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia 19104-6072, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Cengel KA, McKenna WG. Molecular targets for altering radiosensitivity: lessons from Ras as a pre-clinical and clinical model. Crit Rev Oncol Hematol 2005; 55:103-16. [PMID: 16006139 DOI: 10.1016/j.critrevonc.2005.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Revised: 01/01/2005] [Accepted: 02/03/2005] [Indexed: 11/19/2022] Open
Abstract
Ras activation has been correlated with malignant and metastatic cancer phenotypes and poor prognosis for cancer patients. In the preclinical setting, Ras activation by mutation or EGFR amplification results in increased clonogenic cell survival and decreased tumor growth delay following irradiation. Activation of the Ras pathway has also been associated with increased risk of local failure and decreased overall survival in patients receiving radiotherapy. Prenyltransferase inhibitors target the post-translational processing of Ras and have been shown to increase the radiosensitivity of human cancer cell lines. In the clinical setting, these inhibitors have been used with concurrent radiotherapy in a small number of phase I clinical trials with acceptable toxicity. Therefore, inhibiting Ras activation represents a promising molecular approach for radiosensitization in cancer therapy.
Collapse
Affiliation(s)
- Keith A Cengel
- Department of Radiation Oncology, University of Pennsylvania, 3400 Spruce Street, 2 Donner, Philadelphia, PA 19104, USA
| | | |
Collapse
|
19
|
Bourguignon MH, Gisone PA, Perez MR, Michelin S, Dubner D, Giorgio MD, Carosella ED. Genetic and epigenetic features in radiation sensitivity. Eur J Nucl Med Mol Imaging 2005; 32:229-46. [PMID: 15657757 DOI: 10.1007/s00259-004-1730-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent progress especially in the field of gene identification and expression has attracted greater attention to genetic and epigenetic susceptibility to cancer, possibly enhanced by ionising radiation. It has been proposed that the occurrence and severity of the adverse reactions to radiation therapy are also influenced by such genetic susceptibility. This issue is especially important for radiation therapists since hypersensitive patients may suffer from adverse effects in normal tissues following standard radiation therapy, while normally sensitive patients could receive higher doses of radiation offering a better likelihood of cure for malignant tumours. This paper, the first of two parts, reviews the main mechanisms involved in cell response to ionising radiation. DNA repair machinery and cell signalling pathways are considered and their role in radiosensitivity is analysed. The implication of non-targeted and delayed effects in radiosensitivity is also discussed.
Collapse
Affiliation(s)
- Michel H Bourguignon
- Direction Générale de la Sûreté Nucléaire et de la Radioprotection (DGSNR), 6 Place du Colonel Bourgoin, 75572, Paris Cedex 12, France.
| | | | | | | | | | | | | |
Collapse
|
20
|
Affiliation(s)
- Alex A Adjei
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
21
|
Abstract
The combination of radiotherapy and chemotherapy (RCT) has improved the treatment results of patients with inoperable stage-III NSCLC in comparison to irradiation alone and is seen as the standard treatment at present. Concomitant RCT was significantly superior to sequential RCT in three of four studies, so that the simultaneous therapy sequence is seen today as the best modality. Nevertheless, the results, with median survival times of 15-18 months and a 5-year survival rate between 10 and 20%, are still not satisfactory so that further possibilities of optimisation are being looked for: --Can the results of concomitant RCT be improved by an induction chemotherapy or subsequent chemotherapy? --Is there an additional benefit of operative measures for tumours which through the application of RCT get operable? --What is the additional benefit of biological response modifiers applied together with RCT? --Is there a place for RCT of elderly patients or patients in poor general condition? The rationale as well as the initial results shall be discussed in this overview.
Collapse
Affiliation(s)
- Rainer Fietkau
- Department of Radiotherapy of the University of Rostock, Klinik und Poliklinik für Strahlentherapie der Universität Rostock, Südring 75, D- 18059 Rostock, Germany.
| |
Collapse
|
22
|
Abstract
The treatment of squamous cell carcinoma of the head and neck (SCCHN) has recently witnessed the introduction of molecularly targeted agents based on disease biology, target discovery, and validation. One class of agents, the epidermal growth factor receptor (EGFR) inhibitors, is currently in phase III trials. There are multiple processes, however, that appear to be suitable for targeted therapy beyond EGFR. These include signal transduction, cell cycle control, prostaglandin synthesis, protein degradation, hypoxia, and angiogenesis. These systems and specific protein targets will be reviewed in detail with emphasis on promising preclinical and early clinical evidence of activity.
Collapse
Affiliation(s)
- Ezra E W Cohen
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637-1470, USA.
| |
Collapse
|