Genetically Targeted Fractionated Chemotherapy

Read  full  paper  at:http://www.scirp.org/journal/PaperInformation.aspx?PaperID=53967#.VN2pzyzQrzE

Complex and late stage cancer patients are in need of novel methods of selecting and administering chemotherapy particularly for those patients who are refractory to current treatment methods. The use of biomarkers to enhance decision making with regard to the molecular profile of a person’s cancer is becoming more important in the practice of oncology. The standard for the last several decades is to elect chemotherapeutic agents based on staging and histological identification of the primary cancer site alone versus utilizing the genetic and molecular profile information along with histological primary cancer site and staging to select chemotherapy regimens. Cancers are caused by mutations that occur within cells and therefore selecting treatment based on mutations and not primary cancer site alone can provide advantages that may have gone overlooked. As time progresses, more biomarkers continue to be discovered which can lead to more targets for drugs either currently on the market or clinical trials. In addition to advancements made in the progression of cancer treatment with utilizing molecular profiles effectively, there are other therapeutic strategies that have been postulated as advanced effective ways to administer chemotherapy. These strategies provide chemotherapy to patients while fasting, giving insulin or other biological response modifiers adjunctively prior to chemotherapy for enhanced targeting, and giving chemotherapy in micro-doses to allow for increased frequency of administration and the utilization of multiple targeted chemotherapeutic agents concurrently. In this paper we will discuss these topics and explain their benefits in addition to the evidence that supports these treatments. A review on biomarkers and cancer cell metabolism is discussed as it relates to providing a framework for what constitutes a biomarker in addition to what metabolic processes are related to fasting and administering insulin with chemotherapy. The information provided in this document is designed to illuminate and provide evidence for various methodologies that are underserved in the treatment of cancer.

Cite this paper

Smith, A. , Oertle, J. and Prato, D. (2015) Genetically Targeted Fractionated Chemotherapy. Journal of Cancer Therapy, 6, 182-198. doi: 10.4236/jct.2015.62021.

References

[1] Weeks, J.C., Catalano, P.J., Cronin, A., Finkelman, M.D., Mack, J.W., Keating, N.L. and Schrag, D. (2012) Pateints’ Expectation about Effects of Chemotherapy for Advanced Cancer. The New England Journal of Medicine, 367, 1616-1625. http://dx.doi.org/10.1056/NEJMoa1204410
[2] Srinivas, P.R., Kramer, B.S. and Srivastava, S. (2001) Trends in Biomarker Research for Cancer Detection. The Lancet Oncology, 2, 698-704. http://dx.doi.org/10.1016/S1470-2045(01)00560-5
[3] Cho, W.S.C. (2007) Contribution of Oncoproteomics to Cancer Biomarker Discovery. Molecular Cancer, 6, 25. http://dx.doi.org/10.1186/1476-4598-6-25
[4] Hanahan, D. and Weinberg, R.A. (2000) The Hallmarks of Cancer. Cell, 100, 57-70.
http://dx.doi.org/10.1016/S0092-8674(00)81683-9
[5] Ludwig, J.A. and John, N. (2005) Weinstein Biomarkers in Cancer Staging, Prognosis and Treatment Selection. Nature Reviews Cancer, 5, 845-856. http://dx.doi.org/10.1038/nrc1739
[6] Egger, G., Liang, G., Aparicio, A. and Jones, P.A. (2004) Epigenetics in Human Disease and Prospects for Epigenetic Therapy. Nature, 429, 457-463. http://dx.doi.org/10.1038/nature02625
[7] Cristofanilli, M., Budd, G.T., Ellis, M.J., Stopeck, A., Matera, J., Miller, M.C., et al. (2005) Presence of Circulating Tumor Cells (CTC) in Metastatic Breast Cancer (MBC) Predicts Rapid Progression and Poor Prognosis. Journal of Clinical Oncology, 23, 524.
[8] Liyanage, U.K., Moore, T.T., Joo, H.-G., Tanaka, Y., Herrmann, V., Doherty, G., et al. (2002) Prevalence of Regulatory T Cells Is Increased in Peripheral Blood and Tumor Microenvironment of Patients with Pancreas or Breast Adenocarcinoma. Journal of Immunology, 169, 2756-2761.
http://dx.doi.org/10.4049/jimmunol.169.5.2756
[9] Siddiqui, S.A., Frigola, X., Bonne-Annee, S., Mercader, M., Kuntz, S.M., Krambeck, A.E., et al. (2007) Tumor-Infiltrating Foxp3–CD4+CD25+ T Cells Predict Poor Survival in Renal Cell Carcinoma. Clinical Cancer Research, 13, 2075-2081. http://dx.doi.org/10.1158/1078-0432.CCR-06-2139
[10] Woo, E.Y., Yeh, H., Chu, C.S., Schlienger, K., Carroll, R.G., Riley, J.L., et al. (2002) Regulatory T Cells from Lung Cancer Patients Directly Inhibit Autologous T Cell Proliferation. Journal of Immunology, 168, 4272-4276. http://dx.doi.org/10.4049/jimmunol.168.9.4272
[11] Wolf, A.M., Wolf, D., Steurer, M., Gastl, G., Gunsilius, E. and Grubeck-Loebenstein, B. (2003) Increase of Regulatory T Cells in the Peripheral Blood of Cancer Patients. Clinical Cancer Research, 9, 606-612.
[12] Viguier, M., Lema?tre, F., Verola, O., Cho, M.S., Gorochov, G., Dubertret, L., et al. (2004) Foxp3 Expressing CD4+CD25high Regulatory T Cells Are Overrepresented in Human Metastatic Melanoma Lymph Nodes and Inhibit the Function of Infiltrating T Cells. Journal of Immunology, 173, 1444-1453. http://dx.doi.org/10.4049/jimmunol.173.2.1444
[13] Ormandy, L.A., Hillemann, T., Wedemeyer, H., Manns, M.P., Greten, T.F. and Korangy, F. (2005) Increased Populations of Regulatory T Cells in Peripheral Blood of Patients with Hepatocellular Carcinoma. Cancer Research, 65, 2457-1464. http://dx.doi.org/10.1158/0008-5472.CAN-04-3232
[14] Joosten, S.A. and Ottenhoff, T.H.M. (2008) Human CD4 and CD8 Regulatory T Cells in Infectious Diseases and Vaccination. Human Immunology, 69, 760-770. http://dx.doi.org/10.1016/j.humimm.2008.07.017
[15] Bishop, J.M. (1987) The Molecular Genetics of Cancer. Science, 235, 305-311.
http://dx.doi.org/10.1126/science.3541204
[16] Whitfield, M.L., George, L.K., Grant, G.D. and Perou, C.M. (2006) Common Markers of Proliferation. Nature Reviews Cancer, 6, 99-106. http://dx.doi.org/10.1038/nrc1802
[17] Dunning, A.M., Healey, C.S., Pharoah, P.D.P., Teare, D.M., Ponder, B.A.J. and Easton, D.F. (1999) A Systematic Review of Genetic Polymorphisms and Breast Cancer Risk. Cancer Epidemiology, Biomarkers & Prevention, 8, 843-854.
[18] Toru, H., Masaharu, Y., Shinji, T. and Kazuaki, C. (2008) Genetic Polymorphisms and Head and Neck Cancer Risk. International Journal of Oncology, 32, 945-973.
[19] Ignatiadis, M., Xenidis, N., Perraki, M., Apostolaki, S., Politaki, E., Kafousi, M., et al. (2007) Different Prognostic Value of Cytokeratin-19 mRNA Positive Circulating Tumor Cells According to Estrogen Receptor and HER2 Status in Early-Stage Breast Cancer. Journal of Clinical Oncology, 25, 5194-5202.
http://dx.doi.org/10.1200/JCO.2007.11.7762
[20] Delys, L., Detours, V., Franc, B., Thomas, G., Bogdanova, T., Tronko, M., et al. (2007) Gene Expression and the Biological Phenotype of Papillary Thyroid Carcinomas. Oncogene, 26, 7894-7903.
http://dx.doi.org/10.1038/sj.onc.1210588
[21] Dwarakanath, B.S., Manogaran, P.S., Das, S., Das, B.S. and Jain, V. (1994) Heterogeneity in DNA Content and Proliferative Status of Human Brain Tumors. Indian Journal of Medical Research, 100, 279-286.
[22] Weber, J.L. and May, P.E. (1989) Abundant Class of Human DNA Polymorphisms Which Can Be Typed Using Polymerase Chain Reaction. American Journal of Human Genetics, 44, 388-396.
[23] Arzimanoglou, I.I., Gilbert, F. and Barger, H.R. (1998) Microsatellite Instability in Human Solid Tumors. Cancer, 82, 1808-1820.
http://dx.doi.org/10.1002/(SICI)1097-0142(19980515)82:10<1808::AID-CNCR2>3.0.CO;2-J
[24] Arnold, K. (2000) Biomarker for Esophageal Cancer Found in Bloodstream. Journal of the National Cancer Institute, 92, 1787.
[25] Fearnhead, N.S., Britton, M.P. and Bodmer, W.F. (2001) The ABC of APC. Human Molecular Genetics, 10, 721-733. http://dx.doi.org/10.1093/hmg/10.7.721
[26] Kim, H., Kwon, Y.M., Kim, J.S., Lee, H., Park, J.H., Shim, Y.M., et al. (2004) Tumor-Specific Methylation in Bronchial Lavage for the Early Detection of Non-Small-Cell Lung Cancer. Journal of Clinical Oncology, 22, 2363-2370. http://dx.doi.org/10.1200/JCO.2004.10.077
[27] Ehrlich, M. (2002) DNA Methylation in Cancer: Too Much, but Also Too Little. Oncogene, 21, 5400-5413. http://dx.doi.org/10.1038/sj.onc.1205651
[28] Cuezva, J.M., Krajewska, M., Heredia, M.L., Krajewski, S., Kim, G.S.H., Zapata, J.M., et al. (2002) The Bioenergetic Signature of Cancer: A Marker of Tumor Progression. Cancer Research, 62, 6674-6681.
[29] Reivich, M., Kuhl, D., Wolf, A., Greenberg, J., Phelps, M., Ido, T., et al. (1977) Measurement of Local Cerebral Glucose Metabolism in Man with 18F-2-Fluoro-2-deoxy-D-glucose. Acta neurologica Scandinavica. Supplementum, 64, 190-191.
[30] Dichiro, G., Brooks, R.A., Patronas, N.T., Bairamian, D., Kornblith, P.L., Smith, B.H., et al. (1984) Issues in the in Vivo Measurement of Glucose Metabolism of Human Central Nervous System Tumors. Annals of Neurology, 15, 138-146. http://dx.doi.org/10.1002/ana.410150727
[31] Padma, M.V., Said, S., Jacobs, M., Hwang, D.R., Dunigan, K., Satter, M., et al. (2003) Prediction of Pathology and Survival by FDG PET in Gliomas. Journal of Neuro-Oncology, 64, 227-237.
http://dx.doi.org/10.1023/A:1025665820001
[32] Spence, A.M., Muzi, M., Graham, M.M., O’Sullivan, F., Link, J.M., Lewellen, T.K., et al. (2002) 2-[18F]Fluoro-2-deoxyglucose and Glucose Uptake in Malignant Gliomas before and after Radiotherapy: Correlation with Outcome. Clinical Cancer Research, 8, 971-979.
[33] Weber, W.A. (2006) Positron Emission Tomography as an Imaging Biomarker. Journal of Clinical Oncology, 20, 3282-3292. http://dx.doi.org/10.1200/JCO.2006.06.6068
[34] Stricker, T., Vatenacci, D.V.T. and Seiwert, T.Y. (2011) Molecular Profiling of Cancer—The Future of Personalized Cancer Medicine: A Primer on Cancer Biology and the Tools Necessary to Bring Molecular Testing to the Clinic. Seminars in Oncology, 38, 173-185.
http://dx.doi.org/10.1053/j.seminoncol.2011.01.013
[35] Thurlimann, B., Goldhirsch, A., Castiglione, M., Hsu-Schmitz, S.F., Cavalli, F., Bonnefoi, H., et al. (1997) Formestane versus Megestrol Acetate in Postmenopausal Breast Cancer Patients after Failure of Tamoxifen: A Phase III Prospective Randomised Cross over Trial of Second-Line Hormonal Treatment (SAKK 20/90). European Journal of Cancer, 33, 1017-1024.
[36] Stuart, N.S.A., Warwick, J., Blackledge, G.R.P., Spooner, D., Keen, C., Earl, H., et al. (1996) A Randomized Phase III Cross-Over Study of Tamoxifen versus Megestrol Acetate in Advanced and Recurrent Breast Cancer. European Journal of Cancer, 32, 1888-1892.http://dx.doi.org/10.1016/0959-8049(96)00191-8
[37] Cuzick, J. (2007) LHRH-Agonists in Early Breast Cancer Overview Group. Use of Luteinising-Hormone-Releasing Hormone Agonists as Adjuvant Treatment in Premenopausal Patients with Hormone-Receptor-Positive Breast Cancer: A Meta-Analysis of Individual Patient Data from Randomised Adjuvant Trials. The Lancet, 369, 1711-1723. http://dx.doi.org/10.1016/S0140-6736(07)60778-8
[38] Lewis, J.D., Chagpar, A.B., Shaughnessy, E.A., Nurko, J., McMasters, K. and Edwards, M.J. (2010) Excellent Outcomes with Adjuvant Toremifene or Tamoxifen in Early Stage Breast Cancer. Cancer, 116, 2307-2315.
[39] Dowsett, M., Allred, C., Knox, J., Quinn, E., Salter, J., Allred, C., et al. (2008) Relationship between Quantitative Estrogen and Progesterone Receptor Expression and Human Epidermal Growth Factor Receptor 2 (HER-2) Status with Recurrence in the Arimidex, Tamoxifen, Alone or in Combination Trial. Journal of Clinical Oncology, 26, 1059-1065. http://dx.doi.org/10.1200/JCO.2007.12.9437
[40] Bartlett, J.M.S., Rea, D., et al. (2011) Estrogen Receptor and Progesterone Receptor as Predictive Biomarkers of Response to Endocrine Therapy: A Prospectively Powered Pathology Study in the Tamoxifen and Exemestane Adjuvant Multinational Trial. Journal of Clinical Oncology, 29, 1531-1538.
http://dx.doi.org/10.1200/JCO.2010.30.3677
[41] Coombes, R.C., Kilburn, L.S., Snowdon, C.F., Paridaens, R., Coleman, R.E., Bliss, J.M., et al. (2007) Survival and Safety of Exemestane versus Tamoxifen after 2-3 Years’ Tamoxifen Treatment (Intergroup Exemestane Study): A Randomized Controlled Trial. The Lancet, 369, 559-570.
[42] Viale, G., Regan, M.M., Maiorano, E., Mastropasqua, M.G., Golouh, R., Regan, M.M., et al. (2008) Chemoendocrine Compared with Endocrine Adjuvant Therapies for Node-Negative Breast Cancer: Predictive Value of Centrally Reviewed Expression of Estrogen and Progesterone Receptors—International Breast Cancer Study Group. Journal of Clinical Oncology, 26, 1404-1410.
http://dx.doi.org/10.1200/JCO.2007.10.6393
[43] Anderson, H., Hills, M., Zabaglo, L., A’Hern, R., Leary, A.F., Dowsett, M., et al. (2011) Relationship between Estrogen Receptor, Progesterone Receptor, HER-2 and Ki67 Expression and Efficacy of Aromatase Inhibitors in Advanced Breast Cancer. Annals of Oncology, 22, 1770-1776.
http://dx.doi.org/10.1093/annonc/mdq700
[44] Stendahl, M., Ryden, L., Nordenskjold, B., Ebbe J?nsson, P., Landberg, G. and Jirstrom, K. (2006) High Progesterone Receptor Expression Correlates to the Effect of Adjuvant Tamoxifen in Premenopausal Breast Cancer Patients. Clinical Cancer Research, 12, 4614-4618.
http://dx.doi.org/10.1158/1078-0432.CCR-06-0248
[45] Yamashita, H., Ando, Y., Nishio, M., Zhang, Z., Hamaguchi, M., Mita, K., et al. (2006) Immunohisto- chemical Evaluation of Hormone Receptor Status for Predicting Response to Endocrine Therapy in Metastatic Breast Cancer. Breast Cancer, 13, 74-83. http://dx.doi.org/10.2325/jbcs.13.74
[46] Yu, Z., Yang, Q., et al. (2005) Thymidylate Synthase Predicts for Clinical Outcome in Invasive Breast Cancer. Histology and Histopathology, 20, 871-878.
[47] Chen, C.Y., Yang, P.C., et al. (2011) Thymidylate Synthase and Dihydrofolate Reductase Expression in Non-Small Cell Lung Carcinoma: The Association with Treatment Efficacy of Pemetrexed. Lung Cancer, 74, 132-138. http://dx.doi.org/10.1016/j.lungcan.2011.01.024
[48] Lee, S.J., Im, Y.H., et al. (2011) Thymidylate Synthase and Thymidine Phosphorylase as Predictive Markers of Capecitabine Monotherapy in Patients with Anthracycline- and Taxane-Pretreated Metastatic Breast Cancer. Cancer Chemotherapy and Pharmacology, 68, 743-751. http://dx.doi.org/10.1007/s00280-010-1545-0
[49] Penson, R.T., Seiden, M.V., et al. (2004) Expression of Multidrug Resistance-1 Protein Inversely Correlates with Paclitaxel Response and Survival in Ovarian Cancer Patients: A Study in Serial Samples. Gynecologic Oncology, 93, 98-106. http://dx.doi.org/10.1016/j.ygyno.2003.11.053
[50] Yeh, J.J., Hsu, W.H., Wang, J.J., Ho, S.T. and Kao, A. (2003) Predicting Chemotherapy Response to Paclitaxel-Based Therapy in Advanced Non-Small-Cell Lung Cancer with P-Glycoprotein Expression. Respiration, 70, 32-35. http://dx.doi.org/10.1159/000068411
[51] Von Hoff, D.D., Hidalgo, M., et al. (2011) Gemcitabine Plus nab-Paclitaxel Is an Active Regimen in Patients with Advanced Pancreatic Cancer: A Phase I/II Trial. Journal of Clinical Oncology, 29, 4548-4554. http://dx.doi.org/10.1200/JCO.2011.36.5742
[52] Desai, N., Trieu, V., Damascelli, B. and Soon-Shiong, P. (2009) SPARC Expression Correlates with Tumor Response to Albumin-Bound Paclitaxel in Head and Neck Cancer Patients. Translational Oncology, 2, 59-64. http://dx.doi.org/10.1593/tlo.09109
[53] Seve, P., Dumontet, C., et al. (2005) Class III β-Tubulin Expression in Tumor Cells Predicts Response and Outcome in Patients with Non-Small Cell Lung Cancer Receiving Paclitaxel. Molecular Cancer Therapeutics, 4, 2001-2007. http://dx.doi.org/10.1158/1535-7163.MCT-05-0244
[54] Ploussard, G., de la Taille, A., et al. (2010) Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy. Clinical Cancer Research, 70, 9253-9264.
[55] Gao, S., Zhao, X., Lin, B., Hu, Z., Yan, L. and Gao, J. (2012) Clinical Implications of REST and TUBB3 in Ovarian Cancer and Its Relationship to Paclitaxel Resistance. Tumor Biology, 33, 1759-1765. http://dx.doi.org/10.1007/s13277-012-0435-y
[56] Zhang, H.L., Ruan, L., Zheng, L-M., Whyte, D., Tzeng, C-M. and Zhou, X.W. (2012) Association between Class III β-Tubulin Expression and Response to Paclitaxel/Vinorelbine-Based Chemotherapy for Non-Small Cell Lung Cancer: A Meta-Analysis. Lung Cancer, 77, 9-15.
http://dx.doi.org/10.1016/j.lungcan.2012.01.005
[57] Kulkarni, S.A., Ross, D.T., et al. (2009) TLE3 as a Candidate Biomarker of Response to Taxane Therapy. Breast Cancer Research, 11, R17. http://dx.doi.org/10.1186/bcr2241
[58] Gong, W., Dong, J., et al. (2012) RRM1 Expression and Clinical Outcome of Gemcitabine-Containing Chemotherapy for Advanced Non-Small-Cell Lung Cancer: A Meta-Analysis. Lung Cancer, 75, 374-380. http://dx.doi.org/10.1016/j.lungcan.2011.08.003
[59] Ataka, M., Katano, K., et al. (2007) Topoisomerase I Protein Expression and Prognosis of Patients with Colorectal Cancer. Yonago Acta Medica, 50, 81-87.
[60] Braun, M.S., Seymour, M.T., et al. (2008) Predictive Biomarkers of Chemotherapy Efficacy in Colorectal Cancer: Results from the UK MRC FOCUS Trial. Journal of Clinical Oncology, 26, 2690-2698. http://dx.doi.org/10.1200/JCO.2007.15.5580
[61] Kostopoulos, I., Fountzilas, G., et al. (2009) Topoisomerase I but Not Thymidylate Synthase Is Associated with Improved Outcome in Patients with Resected Colorectal Cancer Treated with Irinotecan Containing Adjuvant Chemotherapy. BMC Cancer, 9, 339. http://dx.doi.org/10.1186/1471-2407-9-339
[62] Baselga, J., Swain, S.M., et al. (2012) Pertuzumab Plus Trastumab Plus Docetaxel for Metastatic Breast Cancer. The New England Journal of Medicine, 36, 109-119. http://dx.doi.org/10.1056/NEJMoa1113216
[63] National Comprehensive Cancer Network (2013) NCCN Clinical Practice Guidelines in Oncology. Breast Cancer Version 2.2013.
[64] Bachelot, T., Pujade-Lauraine, E., et al. (2012) Randomized Phase II Trial of Everolimus in Combination with Tamoxifen in Patients with Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Metastatic Breast Cancer with Prior Exposure to Aromatase Inhibitors: A GINECO Study. Journal of Clinical Oncology, 30, 2718-2724 http://dx.doi.org/10.1200/JCO.2011.39.0708
[65] Wolff, A.C., Hayes, D.F., et al. (2013) Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update. Journal of Clinical Oncology, 31, 3997-4013.
http://dx.doi.org/10.1200/JCO.2013.50.9984
[66] Janku, F., Kurzrock, R., et al. (2012) PI3K/AKT/mTOR Inhibitors in Patients with Breast and Gynecologic Malignancies Harboring PIK3CA Mutations. Journal of Clinical Oncology, 30, 777-782.
http://dx.doi.org/10.1200/JCO.2011.36.1196
[67] Janku, F., Kurzrock, R., et al. (2012) PIK3CA Mutation H1047R Is Associated with Response to PI3K/AKT/mTOR Signaling Pathway Inhibitors in Early-Phase Clinical Trials. Cancer Research, 73, 276-284. http://dx.doi.org/10.1158/0008-5472.CAN-12-1726
[68] Moroney, J.W., Kurzrock, R., et al. (2011) A Phase I Trial of Liposomal Doxorubicin, Bevacizumab, and Temsirolimus in Patients with Advanced Gynecologic and Breast Malignancies. Clinical Cancer Research, 17, 6840-6846. http://dx.doi.org/10.1158/1078-0432.CCR-11-0666
[69] Guo, J., Qin, S., et al. (2011) Phase II, Open-Label, Single-Arm Trial of Imatinib Mesylate in Patients with Metastatic Melanoma Harboring c-Kit Mutation or Amplification. Journal of Clinical Oncology, 29, 2904-2909. http://dx.doi.org/10.1200/JCO.2010.33.9275
[70] Carvajal, R.D., Schwartz, G.K., et al. (2011) KIT as a Therapeutic Target in Metastatic Melanoma. Journal of the American Medical Association, 305, 2327-2334. http://dx.doi.org/10.1001/jama.2011.746
[71] Debiec-Rychter, M., Judson, I., et al. (2006) KIT Mutations and Dose Selection for Imatinib in Patients with Advanced Gastrointestinal Stromal Tumours. European Journal of Cancer, 42, 1093-1103.
http://dx.doi.org/10.1016/j.ejca.2006.01.030
[72] Heinrich, M.C., Fletcher, J.A., et al. (2008) Correlation of Kinase Genotype and Clinical Outcome in North American Intergroup Phase III Trial of Imatinib Mesylate for Treatment of Advanced Gastrointestinal Stromal Tumor: CALGB 150105 Study by Cancer and Leukemia Group B and Southwest Oncology Group. Journal of Clinical Oncology, 26, 5360-5367. http://dx.doi.org/10.1200/JCO.2008.17.4284
[73] Cassier, P.A., Hohenberger, P., et al. (2012) Outcome of Patients with Platelet-Derived Growth Factor Receptor Alpha-Mutated Gastrointestinal Stromal Tumors in the Tyrosine Kinase Inhibitor Era. Clinical Cancer Research, 18, 4458-4464. http://dx.doi.org/10.1158/1078-0432.CCR-11-3025
[74] Wells, S.A., Schlumberger, M.J., et al. (2012) Vandetanib in Patients with Locally Advanced or Metastatic Medullary Thyroid Cancer: A Randomized, Double-Blind Phase III Trial. Journal of Clinical Oncology, 30, 134-141. http://dx.doi.org/10.1200/JCO.2011.35.5040
[75] Gennari, A., Bruzzi, P., et al. (2008) HER2 Status and Efficacy of Adjuvant Anthracyclines in Early Breast Cancer: A Pooled Analysis of Randomized Trials. Journal of the National Cancer Institute, 100, 14-20. http://dx.doi.org/10.1093/jnci/djm252
[76] Press, M.F., Slamon, D.J., et al. (2011) Alteration of Topoisomerase II-Alpha Gene in Human Breast Cancer: Association with Responsiveness to Anthracycline Based Chemotherapy. Journal of Clinical Oncology, 29, 859-867. http://dx.doi.org/10.1200/JCO.2009.27.5644
[77] Akimoto, M., Saisho, H., et al. (2006) Relationship between Therapeutic Efficacy of Arterial Infusion Chemotherapy and Expression of P-Glycoprotein and p53 Protein in Advanced Hepatocellular Carcinoma. World Journal of Gastroenterology, 12, 868-873.
[78] Mittal, M.K., Singh, J.P., et al. (2005) Role of p-Glycoprotein Expression in Predicting Response to Neoadjuvant Chemotherapy in Breast Cancer—A Prospective Clinical Study. World Journal of Surgical Oncology, 3, 61. http://dx.doi.org/10.1186/1477-7819-3-61
[79] O’Malley, F.P., Pritchard, K.I., et al. (2011) Topoisomerase II Alpha Protein and Responsiveness of Breast Cancer to Adjuvant Chemotherapy with CEF Compared to CMF in the NCIC CTG Randomized MA.5 Adjuvant Trial. Breast Cancer Research and Treatment, 128, 401-409. http://dx.doi.org/10.1007/s10549-011-1511-5
[80] Rodrigo, R.S., Axel L.C., et al. (2011) Topoisomerase II-Alpha Protein Expression and Histological Response Following Doxorubicin-Based Induction Chemotherapy Predict Survival of Locally Advanced Soft Tissues Sarcomas. European Journal of Cancer, 47, 1319-1327. http://dx.doi.org/10.1016/j.ejca.2011.02.010
[81] Buckingham, L.P., Bonomi, P., et al. (2007) The Prognostic Value of Chromosome 7 Polysomy in Non-Small Cell Lung Cancer Patients Treated with Gefitinib. Journal of Thoracic Oncology, 2, 414-422. http://dx.doi.org/10.1097/01.JTO.0000268675.02744.b0
[82] Verma, S., Blackwell, K., et al. (2012) Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. The New England Journal of Medicine, 367, 1783-1791. http://dx.doi.org/10.1056/NEJMoa1209124
[83] Cortes, J., Baselga, J., et al. (2012) Pertuzumab Monotherapy after Trastuzumab-Based Treatment and Subsequent Reintroduction of Trastuzumab: Activity and Tolerability in Patients with Advanced Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer. Journal of Clinical Oncology, 30, 1594-1600. http://dx.doi.org/10.1200/JCO.2011.37.4207
[84] Bang, Y.J., Kang, Y.K., et al. (2010) Trastuzumab in Combination with Chemotherapy versus Chemotherapy Alone for Treatment of HER2-Positive Advanced Gastric or Gastro-Oesophageal Junction Cancer (ToGA): A Phase 3, Open-Label, Randomised Controlled Trial. The Lancet, 376, 687-697. http://dx.doi.org/10.1016/S0140-6736(10)61121-X
[85] Yin, W., Jiang, Y., Shen, Z., Shao, Z. and Lu, J. (2011) Trastuzumab in the Adjuvant Treatment of HER2-Positive Early Breast Cancer Patients: A Meta-Analysis of Published Randomized Controlled Trials. PLoS ONE, 6, e21030. http://dx.doi.org/10.1371/journal.pone.0021030
[86] Slamon, D., Buyse, M., et al. (2011) Adjuvant Trastuzumab in HER2-Positive Breast Cancer. The New England Journal of Medicine, 365, 1273-1283. http://dx.doi.org/10.1056/NEJMoa0910383
[87] Hurvitz, S.A., Perez, E.A., et al. (2013) Phase II Randomized Study of Trastuzumab Emtansine versus Trastuzumab plus Docetaxel in Patients with Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer. Journal of Clinical Oncology, 31, 1157-1163.
http://dx.doi.org/10.1200/JCO.2012.44.9694
[88] Johnston, S., Pegram, M., et al. (2009) Lapatinib Combined with Letrozole versus Letrozole and Placebo as First-Line Therapy for Postmenopausal Hormone Receptor-Positive Metastatic Breast Cancer. Journal of Clinical Oncology, 27, 5538-5546. http://dx.doi.org/10.1200/JCO.2009.23.3734
[89] Amir, E., Oca?a, A., Seruga, B., Freedman, O. and Clemons, M. (2010) Lapatinib and HER2 Status: Results of a Meta-Analysis of Randomized Phase III Trials in Metastatic Breast Cancer. Cancer Treatment Reviews, 36, 410-415. http://dx.doi.org/10.1016/j.ctrv.2009.12.012
[90] Press, M.F., Finn, R.S., et al. (2008) HER-2 Gene Amplification, HER-2 and Epidermal Growth Factor Receptor mRNA and Protein Expression, and Lapatinib Efficacy in Women with Metastatic Breast Cancer. Clinical Cancer Research, 14, 7861-7870. http://dx.doi.org/10.1158/1078-0432.CCR-08-1056
[91] Chinot, O.L., Barrie, M., et al. (2007) Correlation between O6-Methylguanine-DNA Methyltransferase and Survival in Inoperable Newly Diagnosed Glioblastoma Patients Treated with Neoadjuvant Temozolomide. Journal of Clinical Oncology, 25, 1470-1475. http://dx.doi.org/10.1200/JCO.2006.07.4807
[92] Busch, C., Geislerd, J., Lillehaug, J.R. and Lonning, P.E. (2010) MGMT Expression Levels Predict Disease Stabilisation, Progression-Free and Overall Survival in Patients with Advanced Melanomas Treated with DTIC. European Journal of Cancer, 46, 2127-2133. http://dx.doi.org/10.1016/j.ejca.2010.04.023?
[93] Warburg, O. (1923) Metabolism of Tumours. Biochemische Zeitschrift, 142, 317-333.
[94] Warburg, O. (1925) Iron, the Oxygen-Carrier of Respiration-Ferment. Science, 61, 575-582.
http://dx.doi.org/10.1126/science.61.1588.575
[95] Racker, E. (1972) Bioenergetics and the Problem of Tumor Growth. American Scientist, 60, 56-63.
[96] Turner, J.S. and Brittain, E.G. (1962) Oxygen as a Factor in Photosynthesis. Biological Reviews, 37, 130-170. http://dx.doi.org/10.1111/j.1469-185X.1962.tb01607.x
[97] Lee, C., Raffaghello, L., Brandhorst, S., Safdie, F.M., Bianchi, G., Martin-Montalvo, A., Pistoia, V., Wei, M., Hwang, S., Merlino, A., Emionite, L., Cabo, R. and Longo, V. (2012) Fasting Cycles Retard Growth of Tumor and Sensitize a Range of Cancer Cell Types to Chemotherapy. Science Translational Medicine, 4, 124-127.
[98] Cushman, S.W. and Wardzala, L.J. (1980) Potential Mechanism of Insulin Action on Glucose Transport in the Isolated Rat Adipose Cell. Apparent Translocation of Intracellular Transport Systems to the Plasma Membrane. Journal of Biological Chemistry, 255, 4758-4762.
[99] Suzuki, K. and Kono, T. (1980) Evidence That Insulin Causes Translocation of Glucose Transport Activity to the Plasma Membrane from an Intracellular Storage Site. Proceedings of the National Academy of Sciences of the United States of America, 77, 2542-2545. http://dx.doi.org/10.1073/pnas.77.5.2542
[100] Papa, V., Pezzino, V., Costantino, A., Belfiore, A., Giuffrida, D., Frittitta, L., Vannelli, G.B., Brand, R., Goldfine, I.D. and Vigneri, R. (1990) Elevated Insulin Receptor Content in Human Breast Cancer. Journal of Clinical Investigation, 86, 1503-1510. http://dx.doi.org/10.1172/JCI114868
[101] Belfiore, A. (2007) The Role of Insulin Receptor Isoforms and Hybrid Insulin/IGF-I Receptors in Human Cancer. Current Pharmaceutical Design, 13, 671-686. http://dx.doi.org/10.2174/138161207780249173
[102] Grana, X. and Reddy, E.P. (1995) Cell Cycle Control in Mammalian Cells: Role of Cyclins, Cyclin Dependent Kinases (CDKs), Growth Suppressor Genes and Cyclin-Dependent Kinase Inhibitors (CKIs). Oncogene, 11, 211-219.
[103] Johnson, D.G. and Walker, C.L. (1999) Cyclins and Cell Cycle Checkpoints. Annual Review of Pharmacology and Toxicology, 39, 295-312. http://dx.doi.org/10.1146/annurev.pharmtox.39.1.295
[104] Kaldis, P., Russo, A.A., Chou, H.S., Pavletich, N.P. and Solomon, M.J. (1998) Human and Yeast Cdk-Activating Kinases (CAKs) Display Distinct Substrate Specificities. Molecular Biology of the Cell, 9, 2545-2560. http://dx.doi.org/10.1091/mbc.9.9.2545
[105] Hartwell, L.H. and Kastan, M.B. (1994) Cell Cycle Control and Cancer. Science, 266, 1821-1828. http://dx.doi.org/10.1126/science.7997877
[106] Harper, J. and Elledge, S.J. (1996) Cdk Inhibitors in Development and Cancer. Current Opinion in Genetics & Development, 6, 56-64. http://dx.doi.org/10.1016/S0959-437X(96)90011-8
[107] Senderowicz, A.M. (2002) Cyclin-Dependent Kinases as Targets for Cancer Therapy. In: Giaccone, G., Schilsky, R. and Sondel. P., Eds., Cancer Chemotherapy and Biological Response Modifiers Annual 20, Elsevier Science, New York, 169-188.
[108] Gliozzo, B., Sung, C.K., Scalia, P.L., Papa, V., Frasca, F., Sciacca, L., Giorgino, F., Milazzo, G., Goldfine, I.D., Vigneri, R. and Pezzino, V.J. (1998) Insulin-Stimulated Cell Growth in Insulin Receptor Substrate-1-Deficient ZR-75-1 Cells Is Mediated by a Phosphatidylinositol-3-Kinase-Independent Pathway. Journal of Cellular Biochemistry, 70, 268-280.
http://dx.doi.org/10.1002/(SICI)1097-4644(19980801)70:2<268::AID-JCB12>3.0.CO;2-J
[109] Bentel, J.M., Lebwohl, D.E., Cullen, K.J., Rubin, M.S., Rosen, N., Mendelsohn, J. and Miller, W.H. (1995) Insulin-Like Growth Factors Modulate the Growth Inhibitory Effects of Retinoic Acid on MCF-7 Breast Cancer Cells. Journal of Cellular Physiology, 165, 212-221. http://dx.doi.org/10.1002/jcp.1041650124
[110] Yang, X.F., Beamer, W.G., Huynh, H.T. and Pollak, M. (1996) Cancer Research, 56, 1509-1511.
[111] Pollak, M., Constantino, J., Polychronakos, C., Blauer, S.A., Guyda, H., Redmond, C., Fisher, B. and Margolese, R. (1990) Effect of Tamoxifen on Serum Insulinlike Growth Factor I Levels in Stage I Breast Cancer Patients. Journal of the National Cancer Institute, 82, 1693-1697.
http://dx.doi.org/10.1093/jnci/82.21.1693
[112] Milazzo, G., Giorgino, F., Damante, G., Sung, C., Stampfer, M.R., Vigneri, R., Goldfine, I. and Belfiore, A. (1992) Cancer Research, 52, 3924-3930.
[113] Frittitta, L., Vigneri, R., Stampfer, M.R. and Goldfine, I.D.J. (1995) Insulin Receptor Overexpression in 184B5 Human Mammary Epithelial Cells Induces a Ligand-Dependent Transformed Phenotype. Journal of Cellular Biochemistry, 57, 666-669. http://dx.doi.org/10.1002/jcb.240570411
[114] Kaleko, M., Rutter, W.J. and Miller, A.D. (1990) Molecular and Cellular Biology, 10, 464-473.
[115] Mountjoy, K.G., Finlay, G.J. and Holdaway, I.M. (1987) Cancer Research, 47, 6500-6504.
[116] Benson, E.A. and Holdaway, I.M. (1982) Cancer Research, 42, 1137-1141.
[117] Belfiore, A., Frittitta, L., Costantino, A., Frasca, F., Pandini, G., Sciacca, L., Goldfine, I.D. and Vigneri, R. (1996) Insulin Receptors in Breast Cancer. Annals of the New York Academy of Sciences, 784, 173-188.
http://dx.doi.org/10.1111/j.1749-6632.1996.tb16235.x
[118] Chappell, J., Leitner, W., Solomon, S., Golovchenko, I., Goalstone, M.L. and Draznin, B. (2001) Effect of Insulin on Cell Cycle Progression in MCF-7 Breast Cancer Cells. Journal of Biological Chemistry, 41, 38023-38028.
[119] Reyman, P. (1997) Chemotherapy Principles of Administration. In: Groenwald, S.L., Yarbro, C.H., Goodman, M. and Frogge, M.H., Eds., Cancer Nursing Principles and Practice, 4th Edition.
[120] Dorr, R.T. and Vonn Hoff, D.D. (1993) Cancer Chemotherapy Handbook. 2nd Edition. Appleton & Lange, Norwalk.
[121] Schilsky, R.L., Bailey, B.D. and Chabner, B.A. (1981) Characteristics of Membrane Transport of Methotrexate by Cultured Human Breast Cancer Cells. Biochemical Pharmacology, 30, 1537-1542. http://dx.doi.org/10.1016/0006-2952(81)90378-6
[122] Lippman, M., Bolan, G. and Huff, K. (1976) The Effects of Estrogens and Antiestrogens on Hormone-Responsive Human Breast Cancer in Long-Term Tissue Culture. Cancer Research, 36, 4595-4601.
[123] Monaco, M.E. and Lippman, M.E. (1977) Insulin Stimulation of Fatty Acid Synthesis in Human Breast Cancer in Long Term Tissue Culture. Endocrinology, 101, 1238-1246. http://dx.doi.org/10.1210/endo-101-4-1238
[124] Plich, P.F., Thompson, P.A. and Czech, M.P. (1980) Coordinate Modulation of D-Glucose Transport Activity and Bilayer Fluidity in Plasma Membranes Derived from Control and Insulin-Treated Adipocytes. Proceedings of the National Academy of Sciences of the United States of America, 77, 915-918. http://dx.doi.org/10.1073/pnas.77.2.915
[125] Snyder, A.L., Kann, H.E. and Kohn, K.W. (1971) Inhibition of the Processing of Ribosomal Precursor RNA by Intercalating Agents. Journal of Molecular Biology, 58, 555-565. http://dx.doi.org/10.1016/0022-2836(71)90371-8
[126] Tobey, R.A. (1972) A Simple, Rapid Technique for Determination of the Effects of Chemotherapeutic Agents on Mammalian Cell Cycle Traverse. Cancer Research, 32, 309-316.
[127] Paoletti, C., Lesca, C., Cros, S., Mahy, C. and Auclairs, C. (1979) Ellipticine and Derivateves Induce Breakage of L1210 Cells DNA in Vitro. Biochemical Pharmacology, 28, 345-350.
http://dx.doi.org/10.1016/0006-2952(79)90096-0
[128] Bhuyan, B.K., Faser, T.J. and Li, L.H. (1972) Cell Cycle Phase Specificity and Biochemical Effects of Ellipticine on Mammalian Cells. Cancer Research, 32, 2538-2544.
[129] Saucier, J.M., Festy, B. and Lepecq, J.B. (1971) The Change of Torsion of the DNA Helix Caused by Intercalation. II. Measurement of the Relative Change of Torsion Induced by Various Intercalating Drugs. Biochimie, 53, 973-980. http://dx.doi.org/10.1016/S0300-9084(71)80065-2
[130] Festy, B., Poisson, J. and Paolettic, C. (1975) A New DNA Intercalating Drug: Methoxy-9-Ellipticine to DNA. Cancer Research, 35, 71-76.
[131] Oster, J.B. and Creasey, W.A. (1981) Enhancement of Cellular Uptake of Ellipticine by Insulin Preincubation. European Journal of Cancer and Clinical Oncology, 17, 1097-1103.
http://dx.doi.org/10.1016/0014-2964(81)90294-2
[132] Ohnuma, T. and Holland, J.F. (1997) Nutritional Consequences of Cancer Chemo Therapy and Immunotherapy. Cancer Research, 37, 2395-2406.
[133] Shamberger, R.C., Brennan, M.F., Goodgame, J.T., Lowry, S.F., Maher, M.M., Wesley, R.A. and Pizzo, P.A. (1984) A Prospective Randomized Study of Adjuvant Parenteral Nutrition in the Treatment of Sarcomas: Results of Survival and Metabolic Studies. Surgery, 96, 1-13.
[134] Peacock, J.L., Gorschboth, C.M. and Northon, J.A. (1987) Impact of Insulin on Doxorubicin Induced Rat Host Toxicity and Tumor Regression. Cancer Research, 47, 4318-4322.
[135] Chabner, B.A. and Meyers, C.E. (1985) Clinical Pharmacology of Cancer Chemo Therapy. In: DeVita, V.T., Hellman, S. and Rosenberg, S.A., Eds., Cancer, Principles and Practice of Oncology, J.B. Lippincott Company, Philadelphia, 312.
[136] Kafka, M.S. (1974) The Effect of Insulin on the Permeability of Phosphatidyl Choline Bimolecular Membranes to Glucose. Journal of Membrane Biology, 18, 81-94. http://dx.doi.org/10.1007/BF01870104    eww150213lx

Two Grams BID Is an Oral Dosage of Vitamin C to Reduce the Risk of Recurrence of Superficial Bladder Carcinoma

Read  full  paper  at:http://www.scirp.org/journal/PaperInformation.aspx?PaperID=53879#.VNm_USzQrzE

Background: Continuous exposure to millimolar (mM) Vitamin C (AA) in vitro kills cancer cells. For superficial bladder carcinoma (SBC), standard chemotherapy is instillation of Bacillus Calmette-Guerin. The recurrence rate with this therapy is 91%. But high dosage vitamins including AA reduced the recurrence to 41%. Aim: To determine the oral dosage of AA that causes the highest concentration of AA [AA] in the bladder. Method: We conducted a clinical trial of 14 people who took various dosages of AA, and analyzed the [AA] in their urine. Results: AA above 2 g twice a day was not absorbed. But that intake produced a bladder [AA] above 1 mM in all participants. Conclusion: Taking 2 g of AA BID will increase [AA] in the bladder to a level likely to kill cancer cells that cause SBC. Taking that dosage 2 consecutive days a week is likely to reduce the recurrence rate of SBC substantially.

KEYWORDS

Cancer, Vitamin C, LUTS, Superficial Bladder Carcinoma

Cite this paper

Folk, E. , Downs, T. and Ordman, A. (2015) Two Grams BID Is an Oral Dosage of Vitamin C to Reduce the Risk of Recurrence of Superficial Bladder Carcinoma. Journal of Cancer Therapy, 6, 169-176. doi: 10.4236/jct.2015.62019.

References

[1] Cameron, E. and Pauling, L. (1978) Supplemental Ascorbate in the Supportive Treatment of Cancer: Reevaluation of Prolongation of Survival Times in Terminal Human Cancer. Proceedings of the National Academy of Sciences of the United States of America, 75, 4538-4542.
http://dx.doi.org/10.1073/pnas.75.9.4538
[2] Chen, Q., Levine, M., Buettner, G.R., et al. (2005) Pharmacologic Ascorbic Acid Concentrations Selectively Kill Cancer Cells: Action as a Pro-Drug to Deliver Hydrogen Peroxide to Tissues. Proceedings of the National Academy of Sciences of the United States of America, 102, 13604-13609.
http://dx.doi.org/10.1073/pnas.0506390102
[3] Cameron, E. and Campbell, A. (1974) The Orthomolecular Treatment of Cancer. II. Clinical Trial of High-Dose Ascorbic Acid Supplements in Advanced Human Cancer. Chemico-Biological Interactions, 9, 285-315. http://dx.doi.org/10.1016/0009-2797(74)90019-2
[4] Cameron, E. and Pauling, L. (1976) Supplemental Ascorbate in the Supportive Treatment of Cancer: Prolongation of Survival Times in Terminal Human Cancer. Proceedings of the National Academy of Sciences of the United States of America, 73, 3685-3689. http://dx.doi.org/10.1073/pnas.73.10.3685
[5] Olney, K.E., et al. (2013) Inhibitors of Hydroperoxide Metabolism Enhance Ascorbate-Induced Cytotoxicity. Free Radical Research, 47, 154-163. http://dx.doi.org/10.3109/10715762.2012.755263
[6] Creagan, E.T., et al. (1979) Failure of High-Dose Vitamin C (Ascorbic Acid) Therapy to Benefit Patients with Advanced Cancer: A Controlled Trial. The New England Journal of Medicine, 301, 687-690.
http://dx.doi.org/10.1056/NEJM197909273011303
[7] Moertel, C.G., et al. (1985) High-Dose Vitamin C versus Placebo in the Treatment of Patients with Advanced Cancer That Have Had No Prior Chemotherapy. The New England Journal of Medicine, 312, 137-141. http://dx.doi.org/10.1056/NEJM198501173120301
[8] Padayatty, S.J., et al. (2004) Vitamin C: Intravenous Use by Complementary and Alternative Medicine Practitioners and Adverse Effects. Annals of Internal Medicine, 140, 533-537.
http://dx.doi.org/10.7326/0003-4819-140-7-200404060-00010
[9] Levine, M., et al. (1996) Vitamin C Pharmacokinetics in Healthy Volunteers: Evidence for a Recommended Dietary Allowance. Proceedings of the National Academy of Sciences of the United States of America, 93, 3704-3709. http://dx.doi.org/10.1073/pnas.93.8.3704
[10] Buetter, G.R., et al. (2011) Comment on “Pharmacologic Ascorbate Synergizes with Gemcitabine in Pre-Clinical Models of Pancreatic Cancer” i.e. All We Are Saying Is, Give C a Chance. Free Radical Biology and Medicine, 50, 1726-1727. http://dx.doi.org/10.1016/j.freeradbiomed.2011.03.030
[11] Clinical Trials (2013) http://www.clinicaltrials.gov
[12] National Cancer Institute (2012) Cancer Statistics. http://seer.cancer.gov/statfacts/html/urinb.html
[13] Morales, A. (1980) Treatment of Carcinoma in Situ of the Bladder with BCG. Cancer Immunology, Immunotherapy, 9, 69-72. http://dx.doi.org/10.1007/BF00199531
[14] Lamm, D.L., Riggs, D.R., Shriver, J.S., van Gilder, P.F., Rach, J.F. and De Haven, J.I. (1994) Megadose Vitamins in Bladder Cancer: A Double-Blind Clinical Trial. Journal of Urology, 151, 21-26.
[15] King, G., Beins, M., Larkin, J., Summers, B. and Ordman, A.B. (1994) Rate of Excretion of Vitamin C in Human Urine. AGE, 17, 87-92. http://dx.doi.org/10.1007/BF02435011
[16] Cone, A., Danner, T. and Ordman, A.B. (1996) Urinary Excretion of Calcium in Students and Mature Women Taking Supplements. AGE, 19, 164.
[17] Schleicher, R.L., Carroll, M.D., Ford, E.S. and Lacher, D.A. (2009) Serum Vitamin C and the Prevalence of Vitamin C Deficiency in the United States: 2003-2004 National Health and Nutrition Examination Survey (NHANES). American Journal of Clinical Nutrition, 90, 1252-1263.
http://dx.doi.org/10.3945/ajcn.2008.27016
[18] Moynihan, T. (2012) High-Dose Vitamin C: Can It Kill Cancer Cells? Mayo Clinic Health Information. http://www.mayoclinic.com/health/alternative-cancer-treatment/AN01572
[19] Levine, M., Padayatty, S.J. and Espey, M.G. (2011) Vitamin C: A Concentration-Function Approach Yields Pharmacology and Therapeutic Discoveries. Advances in Nutrition, 2, 78-88.
http://dx.doi.org/10.3945/an.110.000109
[20] Evers, E. (2012) Treat Cancer with IV Vitamin C—Recent Clinical Success. Natural News.
http://www.naturalnews.com/034663_IV_vitamin_c_cancer_treatment.html
[21] Omaye, S.T., Turnbull, J.D. and Sauberlich, H.E. (1979) Selected Methods for the Determination of Ascorbic Acid in Animal Cells. Methods in Enzymology, 62, 3-11.
http://dx.doi.org/10.1016/0076-6879(79)62181-X
[22] Institute of Medicine (2000) Panel on Dietary Antioxidants and Related Compounds, Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. National Academies Press, Washington DC.
[23] Maserejian, N.N., Giovannucci, E.L., McVary, K.T. and McKinlay, J.B. (2011) Dietary, but Not Supplemental, Intakes of Carotenoids and Vitamin C Are Associated with Decreased Odds of Lower Urinary Tract Symptoms in Men. Journal of Nutrition, 141, 267-273. http://dx.doi.org/10.3945/jn.110.132514
[24] Naidu, K.A. (2003) Vitamin C in Human Health and Disease Is Still a Mystery? An Overview. Journal of Nutrition, 2, 7-23. http://dx.doi.org/10.1186/1475-2891-2-7
[25] Thomas, L.D.K., Elinder, C.-G., Tiselius, H.-G., Wolk, A. and Akesson, A. (2013) Ascorbic Acid Supplements and Kidney Stone Incidence among Men: A Prospective Study. JAMA Internal Medicine, 173, 1-2.
[26] Newberry, S.J. (2012) What Is the Evidence That Vitamin C Supplements Lower Blood Pressure? American Journal of Clinical Nutrition, 95, 997-998. http://dx.doi.org/10.3945/ajcn.112.037663
[27] Staff (2012) Vitamin C: Stress Buster. Psychology Today, 25 April 2003.
http://www.psychologytoday.com/articles/200304/vitamin-c-stress-buster
[28] Polidori, M.C., Mecocci, P. and Frei, B. (2001) Plasma Vitamin C Levels Are Decreased and Correlated with Brain Damage in Patients with Intracranial Hemorrhage or Head Trauma. Stroke, 32, 898-902.
http://dx.doi.org/10.1161/01.STR.32.4.898
[29] Polidori, M.C., Praticó, D., Ingegni, T., Mariani, E., Spazzafumo, L. and Sindaco, P.D. (2005) Effects of Vitamin C and Aspirin in Ischemic Stroke-Related Lipid Peroxidation: Results of the AVASAS (Aspirin versus Ascorbic Acid plus Aspirin in Stroke) Study. BioFactors, 24, 265-274.
http://dx.doi.org/10.1002/biof.5520240131
[30] Zweifler, R.M. (2003) Management of Acute Stroke. Southern Medical Journal, 96, 380-385.
http://dx.doi.org/10.1097/01.SMJ.0000063467.75456.7A                      eww150210lx

Cancer, Malnutrition and Cachexia: We Must Break the Triad

Read  full  paper  at:http://www.scirp.org/journal/PaperInformation.aspx?PaperID=53793#.VNMhYizQrzE

Many factors can modify nutritional status in cancer patients, including cachexia, nausea and vomiting, decreased caloric intake or oncologic treatments causing malabsorption. The cachexia-anorexia syndrome is a complex metabolic syndrome associated with cancer and some other palliative conditions characterized by involuntary weight loss involving fat and muscle, anorexia, early satiety, fatigue and weakness due to shifts in metabolism caused by tumour by-products and cytokines. Cachexia is a distressing and debilitating condition, affecting significant numbers of patients with advanced disease and is the primary cause of death in about 20% of all patients with cancer. Though cachexia is most commonly associated with particular tumours, such as head and neck, gastrointestinal tract, pancreas, central nervous system and lung, it may affect any patient with any tumour at any site; no patient and no tumour are excluded. Current treatment for principally depends on its prevention rather than reversing the present disease state, and the clinical results are far from being satisfactory. A careful decision based on good clinical judgement is necessary before deciding to start either enteral or parenteral nutrition, to avoid a useless, costly and difficult treatment. Treatment should be directed toward improvement in the quality of life of the patient and should often include nutritional counseling. It should take into consideration both disease and treatment related factors as well as the cachexia syndrome itself.

Cite this paper

Suhag, V. , Sunita, B. , Sarin, A. and Singh, A. (2015) Cancer, Malnutrition and Cachexia: We Must Break the Triad. International Journal of Medical Physics, Clinical Engineering and Radiation Oncology, 4, 64-70. doi: 10.4236/ijmpcero.2015.41009.

References

[1] Topkan, E., Yavuz, A.A. and Ozyilkan, O. (2007) Cancer Cachexia: Pathophysiologic Aspects and Treatment Options. Asian Pacific Journal of Cancer Prevention, 8, 445-451.
[2] Santarpia, L., Contaldo, F. and Pasanisi, F. (2011) Nutritional Screening and Early Treatment of Malnutrition in Cancer Patients. Journal of Cachexia, Sarcopenia and Muscle, 2, 27-35.
http://dx.doi.org/10.1007/s13539-011-0022-x
[3] Akbulut, G. (2011) New Perspective for Nutritional Support of Cancer Patients: Enteral/Parenteral Nutrition. Experimental and Therapeutic Medicine, 2, 675-684.
[4] Bozzetti, F. (2013) Nutritional Support of the Oncology Patient. Critical Reviews in Oncology & Hematology, 87, 172-200. http://dx.doi.org/10.1016/j.critrevonc.2013.03.006
[5] Bosaeus, I. (2008) Nutritional Support in Multimodal Therapy for Cancer Cachexia. Supportive Care in Cancer, 16, 447-451. http://dx.doi.org/10.1007/s00520-007-0388-7
[6] Bennani-Baiti, N. and Walsh, D. (2009) What Is Cancer Anorexia-Cachexia Syndrome? A Historical Perspective. The Journal of the Royal College of Physicians of Edinburgh, 39, 257-262.
[7] Yavuzsen, T., Walsh, D., Davis, M.P., Kirkova, J., Jin, T., LeGrand, S., et al. (2009) Components of the Anorexia-Cachexia Syndrome: Gastrointestinal Symptom Correlates of Cancer Anorexia. Supportive Care in Cancer, 17, 1531-1541. http://dx.doi.org/10.1007/s00520-009-0623-5
[8] Patra, S.K. and Arora, S. (2012) Integrative Role of Neuropeptides and Cytokines in Cancer Anorexia-Cachexia Syndrome. Clinica Chimica Acta, 413, 1025-1034.
http://dx.doi.org/10.1016/j.cca.2011.12.008
[9] Holmes, S. (2011) Nutrition in the Care of Patients with Cancer Cachexia. British Journal of Community Nursing, 16, 314-318. http://dx.doi.org/10.12968/bjcn.2011.16.7.314
[10] Paccagnella, A., Morassutti, I. and Rosti, G. (2011) Nutritional Intervention for Improving Treatment Tolerance in Cancer Patients. Current Opinion in Oncology, 23, 322-330.
http://dx.doi.org/10.1097/CCO.0b013e3283479c66
[11] Mondello, P., Mian, M., Aloisi, C., Famà, F., Mondello, S. and Pitini, V. (2014) Syndrome: Pathogenesis, Diagnosis, and New Therapeutic Options. Nutrition and Cancer, 16, 1-15.
[12] Penet, M.F., Winnard, P.T., Jacobs, M.A. and Bhujwalla, Z.M. (2011) Understanding Cancer-Induced Cachexia: Imaging the Flame and Its Fuel. Current Opinion in Supportive and Palliative Care, 5, 327-333.
http://dx.doi.org/10.1097/SPC.0b013e32834c49ba
[13] Laviano, A., Meguid, M.M., Inui, A., Muscaritoli, M. and Fanelli, R.F. (2005) Therapy Insight: Cancer Anorexia-Cachexia Syndrome—When All You Can Eat Is Yourself. Nature Clinical Practice Oncology, 2, 158-165.
http://dx.doi.org/10.1038/ncponc0112
[14] Campos-Ferraz, P.L., Andrade, I., Neves, W.D., Hangai, I., Rodrigues-Alves, C.R. and Lancha, A.H. (2014) An Overview of Amines as Nutritional Supplements to Counteract. Journal of Cachexia, Sarcopenia and Muscle, 5, 105-110.
[15] Ockenga, J. and Valentini, L. (2005) Review Article: Anorexia and Cachexia in Gastrointestinal Cancer. Alimentary Pharmacology & Therapeutics, 22, 583-594.
http://dx.doi.org/10.1111/j.1365-2036.2005.02628.x
[16] Barrera, R. (2002) Nutritional Support in Cancer Patients. Journal of Parenteral and Enteral Nutrition, 26, 63-71.
http://dx.doi.org/10.1177/014860710202600516
[17] Bozzetti, F., Mariani, L., Lo Vullo, S., Amerio, M.L., Biffi, R., Caccialanza, G., et al. (2012) The Nutritional Risk in Oncology: A Study of 1,453 Cancer Outpatients. Supportive Care in Cancer, 20, 1919-1928.
http://dx.doi.org/10.1007/s00520-012-1387-x
[18] Bozzetti, F., Arends, J., Lundholm, K., Micklewright, A., Zurcher, G. and Muscaritoli, M. (2009) ESPEN Guidelines on Parenteral Nutrition: Non-Surgical Oncology. Clinical Nutrition, 28, 445-454.
http://dx.doi.org/10.1016/j.clnu.2009.04.011
[19] Bozzetti, F. (2011) Nutritional Support in Oncologic Patients: Where We Are and Where We Are Going. Clinical Nutrition, 30, 714-747. http://dx.doi.org/10.1016/j.clnu.2011.06.011
[20] Penna, F., Minero, V.G., Costamagna, D., Bonelli, G., Baccino, F.M. and Costelli, P. (2010) Anti-Cytokine Strategies for the Treatment of Cancer-Related Anorexia and Cachexia. Expert Opinion on Biological Therapy, 10, 1241-1250.
http://dx.doi.org/10.1517/14712598.2010.503773
[21] Mariani, L., Lo Vullo, S. and Bozzetti, F., on behalf of the SCRINIO Working Group (2011) Weight Loss in Cancer Patients: A Plea for a Better Awareness of the Issue. Supportive Care in Cancer, 20, 301-309.
http://dx.doi.org/10.1007/s00520-010-1075-7
[22] Pascual López, A., Roqué i Figuls, M., Urrútia Cuchi, G., Berenstein, E.G., Almenar Pasies, B., Balcells Alegre, M., et al. (2004) Systematic Review of Megestrol Acetate in the Treatment of Anorexia-Cachexia Syndrome. Journal of Pain and Symptom Management, 27, 360-369. http://dx.doi.org/10.1016/j.jpainsymman.2003.09.007
[23] von Haehling, S. and Anker, S.D. (2014) Treatment of Cachexia: An Overview of Recent Developments. International Journal of Cardiology, 15, 866-872.
[24] Ruiz Garcia, V., López-Briz, E., Carbonel, S.R., Gonzalvez-Perales, J.L. and Bort-Marti, S. (2012) Megestrol Acetate for Treatment of Anorexia-Cachexia Syndrome. The Cochrane Database of Systematic Reviews, 3, CD004310.
[25] Yennurajalingam, S. and Bruera, E. (2014) Role of Corticosteroids for Fatigue in Advanced Incurable Cancer: Is It a “Wonder Drug” or “Deal with the Devil”. Current Opinion in Supportive & Palliative Care, 8, 346-351.
http://dx.doi.org/10.1097/SPC.0000000000000093
[26] Strasser, F. and Bruera, E.D. (2002) Update on Anorexia and Cachexia. Hematology/Oncology Clinics of North America, 16, 589-617. http://dx.doi.org/10.1016/S0889-8588(02)00011-4
[27] Mantovani, G., Madeddu, C. and Macciò, A. (2013) Drugs in Development for Treatment of Patients with Cancer-Related Anorexia and Cachexia Syndrome. Drug Design, Development and Therapy, 7, 645-656.
http://dx.doi.org/10.2147/DDDT.S39771
[28] Balstad, T.R., Kaasa, S. and Solheim, T.S. (2014) Multimodal Nutrition/Anabolic Therapy for Wasting Conditions. Current Opinion in Clinical Nutrition and Metabolic Care, 17, 226-235.
http://dx.doi.org/10.1097/MCO.0000000000000045
[29] Strohle, A., Zanker, K. and Hahn A. (2010) Nutrition in Oncology: The Case of Micronutrients. Oncology Reports, 24, 815-828. http://dx.doi.org/10.3892/or.2010.815
[30] Argilés, J.M., Olivan, M., Busquets, S. and López-Soriano, F.J. (2010) Optimal Management of Cancer Anorexia— Cachexia Syndrome. Cancer Management and Research, 2, 27-38.
http://dx.doi.org/10.2147/CMAR.S7101
[31] Bruera, E. (2013) Emerging Evidence on Psychosocial Effects of Cancer Cachexia: Commentary on Oberholzer et al. Journal of Pain and Symptom Management, 46, 76.
http://dx.doi.org/10.1016/j.jpainsymman.2013.05.005
[32] Bruera, E. and Hui, D. (2013) Palliative Care Research: Lessons Learned by Our Team over the Last 25 Years. Palliative Medicine, 27, 939-951. http://dx.doi.org/10.1177/0269216313477177
[33] Muliawati, Y., Haroen, H. and Rotty, L.W. (2012) Cancer Anorexia-Cachexia Syndrome. Acta Medica Indonesiana, 44, 154-162.
[34] Acreman, S. (2009) Nutrition in Palliative Care. British Journal of Community Nursing, 14, 427-428, 430-431.
http://dx.doi.org/10.12968/bjcn.2009.14.10.44494                                          eww150205lx

The Association of Night Shift Work with the Development of Breast Cancer in Women

Read  full  paper  at:

http://www.scirp.org/journal/PaperInformation.aspx?PaperID=53385#.VMBtkyzQrzE

ABSTRACT

Breast cancer is a serious public health concern in South Africa and globally. It is estimated that one in seven South Africans will develop cancer in their lifetime. According to a case-controlled study, 80% of cancer cases are thought to be due to external, non-inherited factors, which could potentially have been prevented. The objectives of the current case-control study were: 1) to determine the relationship between night shift work and the development of breast cancer; 2) to explore the relationship between night shift work and other types of cancer; 3) to explore any difference between night shift work and breast cancer, and night shift work and other types of cancer. A total of 106 research participants were selected using non-probability, convenience sampling methods and interviewed using a structured questionnaire. Seventy-two (68%) of the women who were interviewed were black, while 32% (n = 35) were white. Of the 106 research participants, 82% (n = 87) had a history of being employed, while 18% (n = 19) had never been employed. Analysed data showed that 29% (n = 31) of the women had a history of working night shift. Of the 31 research participants who reported having worked night shift, 90% (n = 28) had actually done rotating shift work, rather than regular night shift work. The odds ratio of working night shift was found to be 1.24 (OR = 1.24, p = 0.615) higher in breast cancer research participants compared to research participants diagnosed with other types of cancer—odds ratio of 0.80 (p = 0.610). For rotational work, the OR was 1.445, indicating a higher risk than for shift work. It is recommended that the relationship between working night shift and breast cancer risk be explored further through cross-sectional and cohort studies.

Cite this paper

Moukangoe, P. and Jansen van Rensburg, M. (2015) The Association of Night Shift Work with the Development of Breast Cancer in Women. Open Journal of Epidemiology, 5, 14-21. doi: 10.4236/ojepi.2015.51003.

References

[1] Fan, L., Zheng, Y., Yu, K.D., Liu, G.Y., Wu, J., Lu, J.S., Shen, K.W., Shen, Z.Z. and Shao, Z.M. (2009) Breast Cancer in a Transitional Society over 18 Years: Trends and Present Status in Shanghai, China. Breast Cancer Research and Treatment Journal, 117, 409-416.
http://dx.doi.org/10.1007/s10549-008-0303-z
[2] Key, T.J., Verkasalo, P.K. and Banks, E. (2001) Epidemiology of Breast Cancer. Lancet Oncology, 2, 133-140.
http://dx.doi.org/10.1016/S1470-2045(00)00254-0
[3] South African Medical Research Council (2013) What Are the Top Causes of Death Due to Cancer in South Africa? Burden of Disease Research Unit.
http://www.mrc.ac.za/bod/faqcancer.htm
[4] Urban, M., Banks, E., Egger, S., Canfell, K., O’Connell, D., Beral, V. and Sitas, F. (2012) Injectable and Oral Contraceptive Use and Cancers of the Breast, Cervix, Ovary, and Endometrium in Black South African Women: Case-Control Study. PLOS Medicine, 9, 1-11.
[5] Cancer Association of South Africa (2010) Overview of the Latest National Cancer Registry Statistics. Leading Five Female Cancers in South Africa.
http://www.cansa.org.za/womens-health/
[6] Straif, K., Baan, R., Grosse, Y., Secretan, B., Ghissassi, F., Bouvard, V., Altieri, A., Benbrahim-Tallal, L. and Cogliano, V. (2007) Carcinogenicity of Shift-Work, Painting, and Fire-Fighting. Lancet Oncology, 8, 1065-1066.
http://dx.doi.org/10.1016/S1470-2045(07)70373-X
[7] Stevens, R.G. (2009) Light-at-Night, Circadian Disruption and Breast Cancer: Assessment of Existing Evidence. International Journal of Epidemiology, 38, 963-970.
http://dx.doi.org/10.1093/ije/dyp178
[8] Wise, J. (2009) Danish Night Shift Workers with Breast Cancer Awarded Compensation. British Medical Journal, 338, 1152.
[9] Hansen, J. (2006) Risk of Breast Cancer after Night- and Shift Work: Current Evidence and Ongoing Studies in Denmark. Cancer Causes Control, 17, 531-537.
http://dx.doi.org/10.1007/s10552-005-9006-5
[10] Scheer, F.A., Hilton, M.F., Mantzoros, C.S. and Shea, S.A. (2009) Adverse Metabolic and Cardiovascular Consequences of Circardian Misalignment. Proceedings of the National Academy of Science United States of America, 106, 4453-4458.
http://dx.doi.org/10.1073/pnas.0808180106
[11] Costa, G. (2001) Shift-Work Health Consequences. In: Karwowski, W., Eds., International Encyclopedia of Ergonomics and Human Factors, Volume 2, Taylor Francis, London, 1359-1360.
[12] Megdal, S.P., Kroenke, C.H., Laden, F., Pukkala, E. and Schernhammer, E.S. (2005) Night Work and Breast Cancer Risk: A Systematic Review and Meta-Analysis. European Journal of Cancer, 41, 2023-2032.
http://dx.doi.org/10.1016/j.ejca.2005.05.010
[13] Grundy, A., Richardson, H., Burstyn, I., Lohrisch, C., SenGupta, S.K., Lai, A.S., Lee, D., Spinelli, J.J. and Aroson, K.J. (2013) Increased Risk of Breast Cancer Associated with Long-Term Shift Work in CANADA. Occupational and Environmental Medicine, 70, 831-838.
[14] Lie, J.A., Roessink, J. and Kjaerheim, K. (2006) Breast Cancer and Night Work among Norwegian Nurses. Cancer Causes Control, 17, 39-44.
http://dx.doi.org/10.1007/s10552-005-3639-2
[15] Schernhammer, E.S., Laden, F., Speizer, F.E., Willet, W.C., Hunter, D.J., Kawachi, I. and Colditz, G.A. (2001) Rotating Night Shifts and Risk of Breast Cancer in Women Participating in the Nurses’ Health Study. Journal of National Cancer Institute, 93, 1563-1568.
http://dx.doi.org/10.1093/jnci/93.20.1563
[16] Schernhammer, E.S., Kroenke, C.H., Laden, F. and Hankinson, S.E. (2006) Night Work and Risk of Breast Cancer. Epidemiology, 17, 108-111.
http://dx.doi.org/10.1097/01.ede.0000190539.03500.c1
[17] Pesch, B., Harth, V., Rabstein, S., Baish, C., Schiffermann, M., Bonberg, N., Heinze, E., Spickenheuer, A., Justenhoven, C., Brauch, H., Hamann, U., Ko, Y., Straif, K. and Bruning, T. (2010) Night Work and Breast Cancer—Results from the German GENICA Study. Scandanavian Journal of Work, Environment and Health, 36, 134-141.
[18] Anjoeka, P., Bu-Tian, J., Xiao-Ou, S., Shouzheng, X., Gong, Y., Hong-Lan, L., Nathaniel, R., Yu-Tang, G., Wei, Z. and Wong-Ho, Chow. (2010) Night Shift Work and Breast Cancer Risk in a Cohort of Chinese Women. American Journal of Epidemiology, 171, 953-959.
http://dx.doi.org/10.1093/aje/kwq029
[19] Groot, M.T., Baltussen, R., Uyl-de Groot, C.A., Anderson, B.O. and Hortobagyi, G.N. (2005) Costs and Health Effects of Breast Cancer Interventions in Epidemiologically Different Regions of Africa, North America, and Asia. 2nd Biennial Global Summit Consensus Conference on International Breast Health Care, Bethesda, 12-15 January 2005, 581-590.                                                                                         eww150122lx

Challenges in Cancer Care of Elderly

Read  full  paper  at:

http://www.scirp.org/journal/PaperInformation.aspx?PaperID=53346#.VL3NKSzQrzE

ABSTRACT

As a result of vast global improvement of health care and living conditions, the world population is aging. In developed countries, more than half of the cancers occur in patients aged 70 and older. In booming Asian nations, such as India, the aging trend is particularly striking, and therefore geriatric oncology is rapidly coming at the foreground of oncology practice. As these patients have special needs and a different approach to treatment, there is a strong need for the emergence of geriatric oncology as a sub specialty in oncology. Scientific data show that a geriatric assessment identifies many problems in older people with cancer, adds prognostic information, and might improve the outcomes of these patients. There is a genuine unmet requirement to design and implement the following: development of individually tailored geriatric assessment tools for different oncology centers; cooperation of aging and cancer research in the understanding of cancer biology, aging and physiology; improved clinical study designs; development of geriatric oncology programs; and screening tools for geriatric patients made accessible to family physicians.

Cite this paper

Suhag, V. , Sunita, B. , Sarin, A. and Singh, A. (2015) Challenges in Cancer Care of Elderly. International Journal of Medical Physics, Clinical Engineering and Radiation Oncology, 4, 25-31. doi: 10.4236/ijmpcero.2015.41004.

References

[1] Sarkar, A. and Shahi, U.P. (2013) Assessment of Cancer Care in Indian Elderly Cancer Patients: A Single Center Study. South Asian Journal of Cancer, 2, 202-208. http://dx.doi.org/10.4103/2278-330X.119904
[2] Steer, C.B., Marx, G.M. and Singhal, N. (2009) Cancer in Older People: A Tale of Two Disciplines. Internal Medicine Journal, 39, 771-775. http://dx.doi.org/10.1111/j.1445-5994.2009.02056.x
[3] Berger, N.A., Savvides, P. and Koroukian, S.M. (2006) Cancer in the Elderly. Transactions of the American Clinical and Climatological Association, 117, 147-156.
[4] Monfardini, S. (2004) Elderly Oncology: A New Subspecialty? Journal of Clinical Oncology, 22, 4655.
http://dx.doi.org/10.1200/JCO.2004.04.186
[5] Extermann, M. (2010) Geriatric Oncology: An Overview of Progresses and Challenges. Cancer Research and Treatment, 42, 61-68. http://dx.doi.org/10.4143/crt.2010.42.2.61
[6] Hutchins, L.F., Unger, J.M. and Crowley, J.J. (2000) Patients 65 Years of Age or Older in Cancer-Treatment Trials. The New England Journal of Medicine, 342, 1531. http://dx.doi.org/10.1056/NEJM200005183422016
[7] Wildiers, H., Mauer, M. and Pallis, A. (2013) End Points and Trial Design in Geriatric Oncology Research: A Joint European Organisation for Research and Treatment of Cancer—Alliance for Clinical Trials in Oncology. Journal of Clinical Oncology, 31, 3711-3718. http://dx.doi.org/10.1200/JCO.2013.49.6125
[8] Vijaykumar, D.K., Anupama, R. and Gorasia, T.K. (2012) Geriatric Oncology: The Need for a Separate Subspecialty. Indian Journal of Medical Paediatric Oncology, 33, 134-136.
[9] Monfardini, S., Giordano, G. and Sandri, R. (2012) Bringing Geriatrics into Oncology or Also Oncology into Geriatrics? Annals of Oncology, 23, 801. http://dx.doi.org/10.1093/annonc/mdr597
[10] Kunkler, I.H., Audisio, R. and Belkacemi, Y. (2014) Review of Current Best Practice and Priorities for Research in Radiation Oncology for Elderly Patients with Cancer: The International Society of Geriatric Oncology (SIOG) Task Force. Annals of Oncology, 25, 2134-2146. http://dx.doi.org/10.1093/annonc/mdu104
[11] Kenis, C., Schuermans, H. and Van Custem, E. (2009) Screening for a Geriatric Risk Profile in Older Cancer Patients: A Comparative Study of the Predictive Validity of Three Screening Tools. Critical Reviews in Oncology/Hematology, 72, S22. http://dx.doi.org/10.1016/S1040-8428(09)70046-0
[12] Overcash, J.A., Beckstead, J. and Moody, L. (2006) The Abbreviated Comprehensive Geriatric Assessment (aCGA) for Use in the Older Cancer Patient as a Prescreen: Scoring and Interpretation. Critical Reviews in Oncology/Hematology, 59, 205-210. http://dx.doi.org/10.1016/j.critrevonc.2006.04.003
[13] Balducci, L., Lyman, G.H., Ershler, W.B. and Extermann, M. (2004) Comprehensive Geriatric Oncology. 2nd Edition, Taylor & Francis, London & New York.
[14] Extermann, M., Green, T., Tiffenberg, G. and Rich, C.J. (2009) Validation of the Senior Adult Oncology Program (SAOP) 2 Screening Questionnaire. Critical Reviews in Oncology/Hematology, 69, 185.
[15] Monfardini, S., Aapro, M.S. and Bennett, J.M. (2007) Organization of the Clinical Activity of Geriatric Oncology: Report of SIOG (International Society of Geriatric Oncology) Task Force. Critical Reviews in Oncology/Hematology, 62, 62-73. http://dx.doi.org/10.1016/j.critrevonc.2006.10.003
[16] White, H.K. and Cohen, H.J. (2006) The Older Cancer Patient. Medical Clinics of North America, 90, 967-982.
http://dx.doi.org/10.1016/j.mcna.2006.05.017
[17] Pal, S.K., Katheria, V. and Hurria, A. (2010) Evaluating the Older Patient with Cancer: Understanding Frailty and the Geriatric Assessment. CA: A Cancer Journal for Clinicians, 60, 120-132. http://dx.doi.org/10.3322/caac.20059
[18] Surbone, A., Kagawa-Singer, M. and Terret, C. (2007) The Illness Trajectory of Elderly Cancer Patients across Cultures: SIOG Position Paper. Annals of Oncology, 18, 633-638. http://dx.doi.org/10.1093/annonc/mdl178
[19] Anisimov, V.N. (2003) The Relationship between Aging and Carcinogenesis: A Critical Appraisal. Critical Reviews in Oncology/Hematology, 45, 277-304. http://dx.doi.org/10.1016/S1040-8428(02)00121-X
[20] Flood, K.L., Carroll, M.B. and Le, C.V. (2006) Geriatric Syndromes in Elderly Patients Admitted to an Oncology-Acute Care for Elders Unit. Journal of Clinical Oncology, 24, 2298-2303.
http://dx.doi.org/10.1200/JCO.2005.02.8514
[21] Koroukian, S.M., Murray, P. and Madigan, E. (2006) Comorbidity, Disability, and Geriatric Syndromes in Elderly Cancer Patients Receiving Home Health Care. Journal of Clinical Oncology, 24, 2304-2310.
http://dx.doi.org/10.1200/JCO.2005.03.1567
[22] Mohile, S., Dale, W. and Hurria, A. (2012) Geriatric Oncology Research to Improve Clinical Care. Nature Reviews Clinical Oncology, 9, 571-578. http://dx.doi.org/10.1038/nrclinonc.2012.125
[23] Gupta, S.K. and Lamont, E.B. (2004) Patterns of Presentation, Diagnosis, and Treatment in Older Patients with Colon Cancer and Comorbid Dementia. Journal of the American Geriatrics Society, 52, 1681-1687.
http://dx.doi.org/10.1111/j.1532-5415.2004.52461.x
[24] Balducci, L. and Ershler, W.B. (2005) Cancer and Ageing: A Nexus at Several Levels. Nature Reviews Cancer, 5, 655-662. http://dx.doi.org/10.1038/nrc1675
[25] Kumar, A., Soares, H.P. and Balducci, L. (2007) Treatment Tolerance and Efficacy in Elderly Oncology: A Systematic Review of Phase III Randomized Trials Conducted by Five National Cancer Institute-Sponsored Cooperative Groups. Journal of Clinical Oncology, 25, 1272-1276. http://dx.doi.org/10.1200/JCO.2006.09.2759
[26] Caillet, P., Laurent, M. and Bastuji-Garin, S. (2014) Optimal Management of Elderly Cancer Patients: Usefulness of the Comprehensive Geriatric Assessment. Clinical Interventions in Aging, 9, 1645-1660.
[27] Extermann, M. (2003) Studies of Comprehensive Geriatric Assessment in Patients with Cancer. Cancer Control, 10, 463-468.
[28] Puts, M.T., Hardt, J. and Monette, J. (2012) Use of Geriatric Assessment for Older Adults in the Oncology Setting: A Systematic Review. Journal of the National Cancer Institute, 104, 1133-1163.
http://dx.doi.org/10.1093/jnci/djs285
[29] Extermann, M., Aapro, M. and Bernabei, R. (2005) Use of Comprehensive Geriatric Assessment in Older Cancer Patients: Recommendations from the Task Force on CGA of the International Society of Geriatric Oncology (SIOG). Critical Reviews in Oncology/Hematology, 55, 241-252.
http://dx.doi.org/10.1016/j.critrevonc.2005.06.003
[30] Extermann, M. (2010) Evaluation of the Senior Cancer Patient: Comprehensive Geriatric Assessment and Screening Tools for the Elderly. In: Schrijvers, D., Aapro, M., Zakotnik, B., Audisio, R., van Halteren, H. and Hurria, A., Eds., Handbook of Cancer in the Senior Patient, Informa Healthcare, New York, London, 13-21.
http://dx.doi.org/10.3109/9781841847481.003
[31] Hitz, F., Mey, U. and Clough-Gorr, K.M. (2009) Results from a Pilot Study of a Brief Cancer-Specific Geriatric Assessment (CGA) Tool for Use in Clinical Trials in Older Cancer Patients. Critical Reviews in Oncology/Hematology, 72, S20-S21. http://dx.doi.org/10.1016/S1040-8428(09)70044-7
[32] Brighi, N., Balducci, L. and Biasco, G. (2014) Cancer in the Elderly: Is It Time for Palliative Care in Geriatric Oncology? Journal of Geriatric Oncology, 5, 197-203. http://dx.doi.org/10.1016/j.jgo.2014.01.007
[33] Yee, K.W., Pater, J.L. and Pho, L. (2003) Enrollment of Older Patients in Cancer Treatment Trials in Canada: Why Is Age a Barrier? Journal of Clinical Oncology, 21, 1618-1623.
http://dx.doi.org/10.1200/JCO.2003.12.044
[34] Aapro, M.S., Kohne, C.H. and Cohen, J.H. (2005) Never Too Old. Age Should Not Be a Barrier to Enrollment in Cancer Clinical Trials? Oncology, 10, 198-204. http://dx.doi.org/10.1634/theoncologist.10-3-198
[35] Kimmick, G.G., Peterson, B.L., Kornblith, A.B., Mandelblatt, J., Johnson, J.L., Wheeler, J., et al. (2004) Improving Accrual of Older Persons to Cancer Treatment Trials: A Randomized Trial Comparing an Educational Intervention with standard information: CALGB 360001. Journal of Clinical Oncology, 23, 2201-2207.
[36] Kemeny, M.M., Peterson, B.L. and Kornblith, A.B. (2003) Barriers to Clinical Trial Participation by Older Women with Breast Cancer. Journal of Clinical Oncology, 21, 2268-2275.
http://dx.doi.org/10.1200/JCO.2003.09.124
[37] Kornblith, A.B., Kemeny, M., Peterson, B.L., et al. (2002) Survey of Oncologists Perceptions of Barriers to Accrual of Older Patients with Breast Carcinoma to Clinical Trials. Cancer, 95, 989-996.
http://dx.doi.org/10.1002/cncr.10792
[38] Hurria, A., Naylor, M. and Cohen, H.J. (2013) Improving the Quality of Cancer Care in an Aging Population: Recommendations from an IOM Report. The Journal of the American Medical Association, 310, 1795-1796.
http://dx.doi.org/10.1001/jama.2013.280416
[39] Maggiore, R.J., Gorawara-Bhat, R. and Levine, S.K. (2014) Perceptions, Attitudes, and Experiences of Hematology/Oncology Fellows toward Incorporating Geriatrics in Their Training. Journal of Geriatric Oncology, 5, 106-115.
http://dx.doi.org/10.1016/j.jgo.2013.10.003
[40] Williams, G.R., Deal, A.M. and Jolly, T.A. (2014) Feasibility of Geriatric Assessment in Community Oncology Clinics. Journal of Geriatric Oncology, 5, 245-251.
http://dx.doi.org/10.1016/j.jgo.2014.03.001
[41] Kalsi, T., Payne, S. and Brodie, H. (2013) Are the UK Oncology Trainees Adequately Informed about the Needs of Older People with Cancer? British Journal of Cancer, 108, 1936-1941.
http://dx.doi.org/10.1038/bjc.2013.204
[42] Puts, M.T., Santos, B. and Hardt, J. (2014) An Update on a Systematic Review of the Use of Geriatric Assessment for Older Adults in Oncology. Annals of Oncology, 25, 307-315.
http://dx.doi.org/10.1093/annonc/mdt386                                                                     eww150120lx

The Role of Soluble, Insoluble Fibers and Their Bioactive Compounds in Cancer: A Mini Review

Read  full  paper  at:

http://www.scirp.org/journal/PaperInformation.aspx?PaperID=53072#.VLSWf8nQrzE

ABSTRACT

The cancer incidence has risen dramatically over the last decades. About 8 million people died globally according to latest reports, which represented almost 40% more than it was 20 years ago. Risk factors for the development of cancer have been found to include smoking, alcohol, drugs, obesity and diet. Fiber intake has shown to exhibit chemoprotective effects on cancer proliferation and metastasis that may seem to be very promising. This article will review the role of different types of fiber such as, cellulose, lignin, pectin and inulin in development and prevention of different types of cancers. This article would also discuss the effectiveness of both types of fiber in cancer prevention.

Cite this paper

Papandreou, D. , Noor, Z. and Rashed, M. (2015) The Role of Soluble, Insoluble Fibers and Their Bioactive Compounds in Cancer: A Mini Review. Food and Nutrition Sciences, 6, 1-11. doi: 10.4236/fns.2015.61001.

References

[1] Lozano, R., Naghavi, M., Foreman, K., Lim, S., Shibuya, K., Aboyans, V., et al. (2012) Global and Regional Mortality from 235 Causes of Death for 20 Age Groups in 1990 and 2010: A Systematic Analysis for the Global Burden of Disease Study 2010. Lancet, 380, 2095-2128.
http://dx.doi.org/10.1016/S0140-6736(12)61728-0
[2] Jemal, A., Bray, F., Center, M.M., Ferlay, J., Ward, E. and Forman, D. (2011) Global Cancer Statistics. CA: A Cancer Journal for Clinicians, 61, 69-90.
http://dx.doi.org/10.3322/caac.20107
[3] Siegel, R., Ma, J., Zou, Z. and Jemal, A. (2014) Cancer Statistics, 2014. CA: A Cancer Journal for Clinicians, 64, 9-29.
http://dx.doi.org/10.3322/caac.21208
[4] Orang-Ojong, B.B., Munyangaju, J.E., Wei, M.S., Lin, M., Wei, F.G., Foukuang, C. and Zhu, Y. (2013) Impact of Natural Resources and Research on Cancer Treatment and Prevention: A Perspective from Cameroon. Molecular and Clinical Oncology, 1, 610-620.
[5] Zaridze, D., Lifanova, Y., Maximovitch, D., Day, N.E. and Duffy, S.W. (1991) Diet, Alcohol Consumption and Reproductive Factors in a Case-Control Study of Breast Cancer in Moscow. International Journal of Cancer, 48, 493-501.
http://dx.doi.org/10.1002/ijc.2910480404
[6] Sandoval, J. and Esteller, M. (2012) Cancer Epigenomics: Beyond Genomics. Current Opinion in Genetics and Development, 22, 50-55.
http://dx.doi.org/10.1016/j.gde.2012.02.008
[7] Nystrom, M. and Mutanen, M. (2009) Diet and Epigenetics in Colon Cancer. World Journal of Gastroenterology, 15, 257-263.
http://dx.doi.org/10.3748/wjg.15.257
[8] Bannister, A.J. and Kouzarides, T. (2011) Regulation of Chromatin by Histone Modifications. Cell Research, 21, 381395.
http://dx.doi.org/10.1038/cr.2011.22
[9] Murrell, A., Hurd, P.J. and Wood, I.C. (2013) Epigenetic Mechanism in Development and Disease. Biochemical Society Transactions, 41, 697-699.
http://dx.doi.org/10.1042/BST20130051
[10] Buas, M.F. and Vaughan, T.L. (2013) Epidemiology and Risk Factors for Gastroesophageal Junction Tumors: Understanding the Rising Incidence of This Disease. Seminars in Radiation Oncology, 23, 3-9.
http://dx.doi.org/10.1016/j.semradonc.2012.09.008
[11] Geier, M.S., Butler, R.N. and Howarth, G.S. (2006) Probiotics, Prebiotics and Synbiotics: A Role in Chemoprevention for Colorectal Cancer? Cancer Biology and Therapy, 5, 1265-1269.
http://dx.doi.org/10.4161/cbt.5.10.3296
[12] Hutter, C.M., Chang-Claude, J., Slattery, M., Pflugeisen, B.M., Lin, Y., Dugan, D., et al. (2012) Characterization of Gene-Environment Interactions for Colorectal Cancer Susceptibility Loci. Cancer Research, 72, 2036-2044.
http://dx.doi.org/10.1158/0008-5472.CAN-11-4067
[13] Figueiredo, J.C., Hsu, L., Hutter, C.M., Lin, Y., Campbell, P.T., Baron, J.A., et al. (2014) Genome-Wide Diet-Gene Interaction Analyses for Risk of Colorectal Cancer. PLoS Genetics, 10, e1004228.
[14] Park, J.H., Wacholder, S., Gail, M.H., Peters, U., Jacobs, K.B., Chanock, S.J. and Chatterjee, N. (2010) Estimation of Effect Size Distribution from Genome-Wide Association Studies and Implications for Future Discoveries. Nature Genetics, 42, 570-575.
http://dx.doi.org/10.1038/ng.610
[15] Subramaniam, D., Thombre, R., Dhar, A. and Anant, S. (2014) DNA Methyltransferases: A Novel Target for Prevention and Therapy. Frontiers in Oncology, 4, 80.
http://dx.doi.org/10.3389/fonc.2014.00080
[16] Alexander, D.D. and Cushing, C.A. (2011) Red Meat and Colorectal Cancer: A Critical Summary of Prospective Epidemiologic Studies. Obesity Reviews, 12, e472-e493.
http://dx.doi.org/10.1111/j.1467-789X.2010.00785.x
[17] Figueiredo, J.C., Lewinger, J.P., Song, C., Campbell, P.T., Conti, D.V., Edlund, C.K., et al. (2011) Genotype-Environment Interactions in Microsatellite Stable/Microsatellite Instability-Low Colorectal Cancer: Results from a Genome-Wide Association Study. Cancer Epidemiology, Biomarkers and Prevention, 20, 758-766.
http://dx.doi.org/10.1158/1055-9965.EPI-10-0675
[18] Koushik, A., Hunter, D.J., Spiegelman, D., Beeson, W.L., van dan Brandt, P.A., Buring, J.E., et al. (2007) Fruits, Vegetables, and Colon Risk in a Pooled Analysis of 14 Cohort Study. Journal of the National Cancer Institute, 99, 14711483.
http://dx.doi.org/10.1093/jnci/djm155
[19] National Research Council (2005) Dietary Reference Intakes for Energy, Carbohydrate, Fiber, Fat, Fatty Acids, Cholesterol, Protein, and Amino Acids (Macronutrients). The National Academies Press, Washington DC.
[20] Slavin, J.L. (1987) Dietary Fiber: Classification, Chemical Analyses, and Food Sources. Journal of the American Dietetic Association, 87, 1164-1171.
[21] Burkitt, D.P. and Trowell, H.C. (1976) Refined Carbohydrate Foods and Disease—Some Implications of Dietary Fibre. Postgraduate Medical Journal, 52, 476.
http://dx.doi.org/10.1136/pgmj.52.609.476-c
[22] Bravi, F., Scotti, L., Bosetti, C., Bertuccio, P., Negri, E. and La Vecchia, C. (2009) Dietary Fiber and Stomach Cancer Risk: A Case-Control Study from Italy. Cancer Causes and Control, 20, 847-853.
http://dx.doi.org/10.1007/s10552-009-9309-z
[23] Galeone, C., Pelucchi, C., Talamini, R., Negri, E., Montella, M., Ramazzotti, V., et al. (2007) Fibre Intake and Renal Cell Carcinoma: A Case-Control Study from Italy. International Journal of Cancer, 121, 1869-1872.
http://dx.doi.org/10.1002/ijc.22913
[24] Li, H.Q., Diao, Y.T., Li, H., Zhou, Y.Z., Yang, Y.F., Fang, X.Q., Wang, Y., Wu, K., Zhao, D.L., Zhou, R.X. and Lei, F.H. (2007) The Risk Factors Related to Esophageal Squamous Cell Cancer in Feicheng County, China. Chinese Journal of Preventive Medicine, 41, 56-61.
[25] Pelucchi, C., Talamini, R., Galeone, C., Negri, E., Franceschi, S., Dal Maso, L., et al. (2004) Fibre Intake and Prostate Cancer Risk. International Journal of Cancer, 109, 278-280.
http://dx.doi.org/10.1002/ijc.11688
[26] Pelucchi, C., Talamini, R., Levi, F., Bosetti, C., La Vecchia, C., Negri, E., Parpinel, M. and Franceschi, S. (2003) Fibre Intake and Laryngeal Cancer Risk. Annals of Oncology, 14, 162-167.
http://dx.doi.org/10.1093/annonc/mdg032
[27] Soler, M., Bosetti, C., Franceschi, S., Negri, E., Zambon, P., Talamini, R., Conti, E. and La Vecchia, C. (2001) Fiber Intake and the Risk of Oral, Pharyngeal and Esophageal Cancer. International Journal of Cancer, 91, 283-287.
http://dx.doi.org/10.1002/1097-0215(200002)9999:9999&lt;::AID-IJC1047>3.0.CO;2-I
[28] Otles, S. and Ozgoz, S. (2014) Health Effects of Dietary Fiber. Acta Scientiarum Polonorum. Technologia Alimentaria, 13, 191-202.
[29] Papas, M.A., Giovannucci, E. and Platz, E.A. (2004) Fiber from Fruit and Colorectal Neoplasia. Cancer Epidemiology, Biomarkers and Prevention, 13, 1267-1270.
[30] Vanamala, J., Glagolenko, A., Yang, P., Carroll, R.J., Murphy, M.E., Newman, R.A., et al. (2008) Dietary Fish Oil and Pectin Enhance Colonocyte Apoptosis in Part through Suppression of PPARδ/PGE2 and Elevation of PGE3. Carcinogenesis, 29, 790-796.
http://dx.doi.org/10.1093/carcin/bgm256
[31] Spurling, C.C., Suhl, J.A., Boucher, N., Nelson, C.E., Rosenberg, D.W. and Giardina, C. (2008) The Short Chain Fatty Acid Butyrate Induces Promoter Demethylation and Reactivation of RARβ2 in Colon Cancer Cells. Nutrition and Cancer, 60, 692-702.
http://dx.doi.org/10.1080/01635580802008278
[32] Bidoli, E., Pelucchi, C., Polesel, J., Negri, E., Barzan, L., Franchin, G., Franceschi, S., Serraino, D., De Paoli, P., La Vecchia, C. and Talamini, R. (2013) Fiber Intake and Risk of Nasopharyngeal Carcinoma: A Case-Control Study. Nutrition and Cancer, 65, 1157-1163.
http://dx.doi.org/10.1080/01635581.2013.828088
[33] Daniel, C.R., Park, Y., Chow, W.H., Graubard, B.I., Hollenbeck, A.R. and Sinha, R. (2013) Intake of Fiber and FiberRich Plant Foods Is Associated with a Lower Risk of Renal Cell Carcinoma in a Large US Cohort. The American Journal of Clinical Nutrition, 97, 1036-1043.
http://dx.doi.org/10.3945/ajcn.112.045351
[34] Goodman, M.T., Wilkens, L.R., Hankin, J.H., Lyu, L.C., Wu, A.H. and Kolonel, L.N. (1997) Association of Soy and Fiber Consumption with the Risk of Endometrial Cancer. American Journal of Epidemiology, 146, 294-306.
http://dx.doi.org/10.1093/oxfordjournals.aje.a009270
[35] Jacobs, L.R. (1986) Relationship between Dietary Fiber and Cancer: Metabolic, Physiologic, and Cellular Mechanisms. Experimental Biology and Medicine, 183, 299-310.
http://dx.doi.org/10.3181/00379727-183-42423
[36] Jacobs, L.R. (1986) Modification of Experimental Colon Carcinogenesis by Dietary Fibers. Advances in Experimental Medicine and Biology, 206, 105-118.
[37] Madar, Z., Gurevich, P., Ben-Hur, H., Ben-Arie, A., Berman, V., Sandler, B., et al. (1998) Effects of Dietary Fiber on the Rat Intestinal Mucosa Exposed to Low Doses of a Carcinogen. Anticancer Research, 18, 3521-3526.
[38] Goncalves, P. and Martel, F. (2013) Butyrate and Colorectal Cancer: The Role of Butyrate Transport. Current Drug Metabolism, 14, 994-1008.
http://dx.doi.org/10.2174/1389200211314090006
[39] Kritchevsky, D. (1997) Dietary Fibre and Cancer. European Journal of Cancer Prevention, 6, 435-441.
http://dx.doi.org/10.1097/00008469-199710000-00004
[40] Cappellani, A., Zanghi, A., Di Vita, M., Cavallaro, A., Piccolo, G., Veroux, P., et al. (2013) Strong Correlation between Diet and Development of Colorectal Cancer. Frontiers in Bioscience, 18, 190-198.
http://dx.doi.org/10.2741/4095
[41] Ferguson, L.R., Roberton, A.M., Watson, M.E., Kestell, P. and Harris, P.J. (1993) The Adsorption of a Range of Dietary Carcinogens by α-Cellulose, a Model Insoluble Dietary Fiber. Mutation Research, 319, 257-266.
http://dx.doi.org/10.1016/0165-1218(93)90013-4
[42] Kaczmarczyk, M.M., Miller, M.J. and Freund, G.G. (2012) The Health Benefits of Dietary Fiber: Beyond the Usual Suspects of Type 2 Diabetes Mellitus, Cardiovascular Disease and Colon Cancer. Metabolism-Clinical and Experimental, 61, 1058-1066.
http://dx.doi.org/10.1016/j.metabol.2012.01.017
[43] Fiorucci, S., Cipriani, S., Mencarelli, A., Renga, B., Distrutti, E. and Baldelli, F. (2010) Counter-Regulatory Role of Bile Acid Activated Receptors in Immunity and Inflammation. Current Molecular Medicine, 10, 579-595.
[44] Floch, M.H., Wolfman, M. and Doyle, R. (1980) Fiber and Gastrointestinal Microecology. Journal of Clinical Gastroenterology, 2, 175-184.
http://dx.doi.org/10.1097/00004836-198006000-00014
[45] Kay, R.M. (1982) Dietary Fiber. Journal of Lipid Research, 23, 221-242.
[46] Cheah, P.Y. and Bernstein, H. (1990) Colon Cancer and Dietary Fiber: Cellulose Inhibits the DNA-Damaging Ability of Bile Acids. Nutrition and Cancer, 13, 51-57.
http://dx.doi.org/10.1080/01635589009514044
[47] Ajouz, H., Mukherji, D. and Shamseddine, A. (2014) Secondary Bile Acids: An Underrecognized Cause of Colon Cancer. World Journal of Surgical Oncology, 12, 164.
http://dx.doi.org/10.1186/1477-7819-12-164
[48] Hofmann, A.F. and Rods, A.A. (1984) Physicochemical Properties of Bile Acids and Their Relationship to Biological Properties: An Overview of the Problem. Journal of Lipid Research, 25, 1477-1489.
[49] Degirolamo, C., Modica, S., Palasciano, G. and Moschetta, A. (2011) Bile Acids and Colon Cancer: Solving the Puzzle with Nuclear Receptors. Trends in Molecular Medicine, 17, 564-572.
http://dx.doi.org/10.1016/j.molmed.2011.05.010
[50] Sloan, D.A., Fleiszer, D.M., Richards, G.K., Murray, D. and Brown, R.A. (1993) The Effect of the Fiber Components Cellulose and Lignin on Experimental Colon Neoplasia. Journal of Surgical Oncology, 52, 77-82.
http://dx.doi.org/10.1002/jso.2930520204
[51] Pelucchi, C., La Vecchia, C., Chatenoud, L., Negri, E., Conti, E., Montella, M., Calza, S., Dal Maso, L. and Franceschi, S. (2001) Dietary Fibres and Ovarian Cancer Risk. European Journal of Cancer, 37, 2235-2239.
http://dx.doi.org/10.1016/S0959-8049(01)00291-X
[52] La Vecchia, C., Ferraroni, M., Franceschi, S., Mezzetti, M., Decarli, A. and Negri, E. (1997) Fibers and Breast Cancer Risk. Nutrition and Cancer, 28, 264-269.
http://dx.doi.org/10.1080/01635589709514586
[53] Lee, S., Monnappa, A.K. and Mitchell, R.J. (2012) Biological Activities of Lignin Hydrolysate-Related Compounds. BMB Reports, 45, 265-274.
http://dx.doi.org/10.5483/BMBRep.2012.45.5.265
[54] Lu, F.J., Chu, L.H. and Gau, R.J. (1998) Free Radical-Scavenging Properties of Lignin. Nutrition and Cancer, 30, 3138.
http://dx.doi.org/10.1080/01635589809514637
[55] Ferguson, L.R. and Harris, P.J. (1996) Studies on the Role of Specific Dietary Fibres in Protection against Colorectal Cancer. Mutation Research, 350, 173-184.
http://dx.doi.org/10.1016/0027-5107(95)00105-0
[56] Freeman, H.J. (1979) Dietary Fibre and Colonic Neoplasia. Canadian Medical Association Journal, 121, 291-296.
[57] McPherson-Kay, R. (1987) Fiber, Stool Bulk, and Bile Acid Output: Implications for Colon Cancer Risk. Preventive Medicine, 16, 540-544.
http://dx.doi.org/10.1016/0091-7435(87)90069-7
[58] Adlercreutz, H. (1990) Western Diet and Western Diseases: Some Hormonal and Biochemical Mechanisms and Associations. Scandinavian Journal of Clinical and Laboratory Investigation. Supplementum, 50, 3-23.
http://dx.doi.org/10.3109/00365519009085798
[59] Reddy, B.S., Maeura, Y. and Wayman, M. (1983) Effect of Dietary Corn Bran and Autohydrolyzed Lignin on 3,2’Dimethyl-4-Aminobiphenyl-Induced Intestinal Carcinogenesis in Male F344 Rats. Journal of the National Cancer Institute, 71, 419-423.
[60] Adlercreutz, H., Honjo, H., Higashi, A., Fotsis, T., Hamalainen, E., Hasegawa, T. and Okada, H. (1991) Urinary Excretion of Lignans and Isoflavonoid Phytoestrogens in Japanese Men and Women Consuming a Traditional Japanese Diet. The American Journal of Clinical Nutrition, 54, 1093-1100.
[61] Buck, K., Zaineddin, A.K., Vrieling, A., Heinz, J., Linseisen, J., Flesch-Janys, D. and Chang-Claude, J. (2011) Estimated Enterolignans, Lignan-Rich Foods, and Fibre in Relation to Survival after Postmenopausal Breast Cancer. British Journal of Cancer, 105, 1151-1157.
http://dx.doi.org/10.1038/bjc.2011.374
[62] Saarinen, N.M., Warri, A., Airio, M., Smeds, A. and Makela, S. (2007) Role of Dietary Lignans in the Reduction of Breast Cancer Risk. Molecular Nutrition and Food Research, 51, 857-866.
http://dx.doi.org/10.1002/mnfr.200600240
[63] Adlercreutz, H., Mousavi, Y., Clark, J., Hockerstedt, K., Hamalainen, E., Wahala, K., Makela, T. and Hase, T. (1992) Dietary Phytoestrogens and Cancer: In Vitro and in Vivo Studies. The Journal of Steroid Biochemistry and Molecular Biology, 41, 331-337.
http://dx.doi.org/10.1016/0960-0760(92)90359-Q
[64] Horn-Ross, P.L., Hoggatt, K.J. and Lee, M.M. (2002) Phytoestrogens and Thyroid Cancer Risk: The San Francisco Bay Area Thyroid Cancer Study. Cancer Epidemiology, Biomarkers and Prevention, 11, 43-49.
[65] Leclere, L., Custem, P.V. and Michiels, C. (2013) Anti-Cancer Activities of pHor Heat-Modified Pectin. Frontiers in Pharmacology, 4, 128.
http://dx.doi.org/10.3389/fphar.2013.00128
[66] Barry, K.A., Wojcicki, B.J., Middelbos, I.S., Vester, B.M., Swanson, K.S. and Fahey Jr., G.C. (2010) Dietary Cellulose, Fructooligosaccharides, and Pectin Modify Fecal Protein Catabolites and Microbial Populations in Adult Cats. Journal of Animal Science, 88, 2978-2987.
http://dx.doi.org/10.2527/jas.2009-2464
[67] Attari, F., Sephehri, H., Delphi, L. and Golliaei, B. (2009) Apoptotic and Necrotic Effects of Pectic Acid on Rat Pituitary GH3/B6 Tumor Cells. Iranian Biomedical Journal, 13, 229-236.
[68] Cho, Y., Turner, N.D., Davidson, L.A., Chapkin, R.S., Carroll, R.J. and Lupton, J.R. (2012) A Chemoprotective Fish Oil/Pectin Diet Enhances Apoptosis via Bcl-2 Promoter Methylation in Rat Azoxymethane-Induced Carcinomas. Experimental Biology and Medicine (Maywood, N.J.), 237, 1387-1393.
http://dx.doi.org/10.1258/ebm.2012.012244
[69] Hao, M., Yuan, X., Cheng, H., Xue, H., Zhang, T., Zhou, Y. and Tai, G. (2013) Comparative Studies on the Anti Tumor Activities of High Temperatureand pH-Modified Citrus Pectins. Food and Function, 4, 960-971.
http://dx.doi.org/10.1039/c3fo30350k
[70] Liu, Y., Colditz, G.A., Cotterchio, M., Boucher, B.A. and Kreiger, N. (2014) Adolescent Dietary Fiber, Vegetable Fat, Vegetable Protein, and Nut Intakes and Breast Cancer Risk. Breast Cancer Research and Treatment, 145, 461-470.
http://dx.doi.org/10.1007/s10549-014-2953-3
[71] Hsu, D.K., Chen, H.Y. and Liu, F.T. (2009) Galectin-3 Regulates T-Cell Functions. Immunological Reviews, 230, 114127.
http://dx.doi.org/10.1111/j.1600-065X.2009.00798.x
[72] Nagina-Makker, P., Hogan, V., Honjo, Y., Baccarini, S., Tait, L., Bresalier, R. and Raz, A. (2002) Inhibition of Human Cancer Cell Growth and Metastasis in Nude Mice by Oral Intake of Modified Citrus Pectin. Journal of the National Cancer Institute, 94, 1854-1862.
http://dx.doi.org/10.1093/jnci/94.24.1854
[73] Zeng, H. and Briske-Anderson, M. (2005) Prolonged Butyrate Treatment Inhibits the Migration and Invasion Potential of HT1080 Tumor Cells. Journal of Nutrition, 135, 291-295.
[74] Elangovan, S., Pathania, R., Ramchandran, S., Ananth, S., Padia, R.N., Lan, L., et al. (2014) The Niacin/Butyrate Receptor GPR109A Suppresses Mammary Tumorigenesis by Inhibiting Cell Survival. Cancer Research, 74, 1166-1178.
http://dx.doi.org/10.1158/0008-5472.CAN-13-1451
[75] Inohara, H. and Raz, A. (1994) Effects of Natural Complex Carbohydrate (Citrus Pectin) on Murine Melanoma Cell Properties Related to Galectin-3 Functions. Glycoconjugate Journal, 11, 527-532.
http://dx.doi.org/10.1007/BF00731303
[76] Platt, D. and Raz, A. (1992) Modulation of the Lung Colonization of B16-F1 Melanoma Cells by Citrus Pectin. Journal of the National Cancer Institute, 84, 438-442.
http://dx.doi.org/10.1093/jnci/84.6.438
[77] Hague, A., Elder, D.J., Hicks, D.J. and Paraskeva, C. (1995) Apoptosis in Colorectal Tumour Cells: Induction by the Short Chain Fatty Acids Butyrate, Propionate and Acetate and by the Bile Salt Deoxycholate. International Journal of Cancer, 60, 400-406.
http://dx.doi.org/10.1002/ijc.2910600322
[78] Xiao, M., Liu, Y.G., Zou, M.C. and Zou, F. (2014) Sodium Butyrate Induces Apoptosis of Human Colon Cancer Cells by Modulating ERK and Sphingosine Kinase 2. Biomedical and Environmental Sciences, 27, 197-203.
[79] Hague, A., Manning, A.M., Hanlon, K.A., Huschtscha, L.I., Hart, D. and Paraskeva, C. (1993) Sodium Butyrate Induces Apoptosis in Human Colonic Tumour Cell Lines in a p53-Independent Pathway: Implications for the Possible Role of Dietary Fibre in the Prevention of Large-Bowel Cancer. International Journal of Cancer, 55, 498-505.
http://dx.doi.org/10.1002/ijc.2910550329
[80] Roberfroid, M.B. (2005) Introducing Inulin-Type Fructans. British Journal of Nutrition, 93, S13-S25.
http://dx.doi.org/10.1079/BJN20041350
[81] Pasqualetti, V., Altomare, A., Guarino, M.P., Locato, V., Cocca, S., Cimini, S., Palma, R., Alloni, R., De Gara, L. and Cicala, M. (2014) Antioxidant Activity of Inulin and Its Role in the Prevention of Human Colonic Muscle Cell Impairment Induced by Lipopolysaccharide Mucosal Exposure. PLoS ONE, 9, e98031.
http://dx.doi.org/10.1371/journal.pone.0098031
[82] Burns, A.J. and Rowland, I.R. (2000) Anti-Carcinogenicity of Probiotics and Prebiotics. Current Issues in Intestinal Microbiology, 1, 13-24.
[83] Taper, H.S. and Roberfroid, M.B. (2000) Inhibitory Effect of Dietary Inulin or Oligofructose on the Development of Cancer Metastases. Anticancer Research, 20, 4291-4294.                                                eww150113lx

Chlorambucil Encapsulation into PLGA Nanoparticles and Cytotoxic Effects in Breast Cancer Cell

Read  full  paper  at:

http://www.scirp.org/journal/PaperInformation.aspx?PaperID=53000#.VK9BqsnQrzE

ABSTRACT

The present work aimed to develop and evaluate a colloidal system composed of poly (DL-lactide-co-glycolide) (PLGA) nanoparticles (NPs) associated with chlorambucil (CHB) and its effects on cancer cells. The nanoparticles showed %EE (>92%), a mean particle size in the range of 240 to 334 nm and zeta potential of -16.7 to -26.0 mV. In vitro release profile showed a biphasic pattern, with an initial burst for all formulations. The scanning electron microscopy of CHB-nanoparticles showed regular spherical shapes, smooth surface without aggregations. Differential scanning calorimetry thermograms, UV-vis absorption, fluorescence emission and Fourier transform infrared spectroscopy were performed showing the entrapment of the antitumoral in drug delivery system. CHB encapsulated in PLGA nanoparticles decrease the survival rates of the breast cancer cells: 68.9% reduction of cell viability on MCF-7 cell line and 59.7% on NIH3T3. Our results indicated that polymeric nanoparticles produced by classical methods are efficient drug delivery systems for CHB.

Cite this paper

Dias, D. , Joanitti, G. , Azevedo, R. , Silva, L. , Lunardi, C. and Gomes, A. (2015) Chlorambucil Encapsulation into PLGA Nanoparticles and Cytotoxic Effects in Breast Cancer Cell. Journal of Biophysical Chemistry, 6, 1-13. doi: 10.4236/jbpc.2015.61001.

References

[1] Zhang, H., Ye, Z.W. and Lou, Y.J. (2004) Metabolism of Chlorambucil by Rat Liver Microsomal Glutathione S-Transferase. Chemico-Biological Interactions, 149, 61-67.
http://dx.doi.org/10.1016/j.cbi.2003.07.002
[2] Schmidt, M. (2014) Chemotherapy in Early Breast Cancer: When, How and Which One? Breast Care (Basel), 9, 154- 160.
http://dx.doi.org/10.1159/000363755
[3] Cummings, S.R., Tice, J.A., Bauer, S., Browner, W.S., Cuzick, J., Ziv, E., Vogel, V., Shepherd, J., Vachon, C., Smith- Bindman, R. and Kerlikowske, K. (2009) Prevention of Breast Cancer in Postmenopausal Women: Approaches to Estimating and Reducing Risk. Journal of the National Cancer Institute, 101, 384-398.
http://dx.doi.org/10.1093/jnci/djp018
[4] Zhang, W.J., Feng, M.L., Zheng, G.P., Chen, Y., Wang, X.D., Pen, B., Yin, J., Yu, Y.H. and He, Z.M. (2012) Chemoresistance to 5-Fluorouracil Induces Epithelial-Mesenchymal Transition via Up-Regulation of Snail in MCF7 Human Breast Cancer Cells. Biochemical and Biophysical Research Communications, 417, 679-685.
http://dx.doi.org/10.1016/j.bbrc.2011.11.142
[5] Bergh, J., Jonsson, P.E., Glimelius, B., Nygren, P. and Care, S.B.-G.S.C.O.T.A.I.H. (2001) A Systematic Overview of Chemotherapy Effects in Breast Cancer. Acta Oncologica, 40, 253-281.
http://dx.doi.org/10.1080/02841860151116349
[6] Anderson, W.F., Rosenberg, P.S., Prat, A., Perou, C.M. and Sherman, M.E. (2014) How Many Etiological Subtypes of Breast Cancer: Two, Three, Four, or More? Journal of the National Cancer Institute, 106, 1-11.
[7] Pettersson, A., Graff, R.E., Ursin, G., Santos Silva, I.D., McCormack, V., Baglietto, L., Vachon, C., Bakker, M.F., Giles, G.G., Chia, K.S., Czene, K., Eriksson, L., Hall, P., Hartman, M., Warren, R.M., Hislop, G., Chiarelli, A.M., Hopper, J.L., Krishnan, K., Li, J., Li, Q., Pagano, I., Rosner, B.A., Wong, C.S., Scott, C., Stone, J., Maskarinec, G., Boyd, N.F., van Gils, C.H. and Tamimi, R.M. (2014) Mammographic Density Phenotypes and Risk of Breast Cancer: A Meta-Analysis. Journal of the National Cancer Institute, 106, 1-11.
[8] Bielawski, K., Bielawska, A., Muszynska, A., Poplawska, B. and Czarnomysy, R. (2011) Cytotoxic Activity of G3 PAMAM-NH2 Dendrimer-Chlorambucil Conjugate in Human Breast Cancer Cells. Environmental Toxicology and Pharmacology, 32, 364-372.
http://dx.doi.org/10.1016/j.etap.2011.08.002
[9] Benitah, N., de Lorimier, L.-P., Gaspar, M. and Kitchell, B.E. (2003) Chlorambucil-Induced Myoclonus in a Cat with Lymphoma. Journal of the American Animal Hospital Association, 39, 283-287.
http://dx.doi.org/10.5326/0390283
[10] Hehn, S.T., Dorr, R.T. and Miller, T.P. (2003) Mood Alterations in Patients Treated with Chlorambucil. Clinical lymphoma, 4, 179-182.
http://dx.doi.org/10.3816/CLM.2003.n.028
[11] Reux, B., Weber, V., Galmier, M.J., Borel, M., Madesclaire, M., Madelmont, J.C., Debiton, E. and Coudert, P. (2008) Synthesis and Cytotoxic Properties of New Fluorodeoxyglucose-Coupled Chlorambucil Derivatives. Bioorganic & Medicinal Chemistry, 16, 5004-5020.
http://dx.doi.org/10.1016/j.bmc.2008.03.038
[12] Ganta, S., Paxton, J.W., Baguley, B.C. and Garg, S. (2008) Pharmacokinetics and Pharmacodynamics of Chlorambucil Delivered in Parenteral Emulsion. International Journal of Pharmaceutics, 360, 115-121.
http://dx.doi.org/10.1016/j.ijpharm.2008.04.027
[13] Yordanov, G.G., Bedzhova, Z.A. and Dushkin, C.D. (2010) Preparation and Physicochemical Characterization of Novel Chlorambucil-Loaded Nanoparticles of Poly(Butylcyanoacrylate). Colloid and Polymer Science, 288, 893-899.
http://dx.doi.org/10.1007/s00396-010-2219-5
[14] Descoteaux, C., Leblanc, V., Brasseur, K., Gupta, A., Asselin, E. and Berube, G. (2010) Synthesis of D-and L-Tyrosine- Chlorambucil Analogs Active against Breast Cancer Cell Lines. Bioorganic & Medicinal Chemistry Letters, 20, 7388- 7392.
http://dx.doi.org/10.1016/j.bmcl.2010.10.039
[15] Omoomi, F.D. (2013) Molecular Chlorambucil-Methionine Conjugate: Novel Anti-Cancer Agent against Breast MCF-7 Cell Model. Journal of Cancer Science & Therapy, 5, 75-84.
[16] Millard, M., Gallagher, J.D., Olenyuk, B.Z. and Neamati, N. (2013) A Selective Mitochondrial-Targeted Chlorambucil with Remarkable Cytotoxicity in Breast and Pancreatic Cancers. Journal of Medicinal Chemistry, 56, 9170-9179.
http://dx.doi.org/10.1021/jm4012438
[17] Gomes, A.J., Barbougli, P.A., Espreafico, E.M. and Tfouni, E. (2008) Trans-[Ru(NO)(NH3)4(py)](BF4)3·H2O Encapsulated in PLGA Microparticles for Delivery of Nitric Oxide to B16-F10 Cells: Cytotoxicity and Phototoxicity. Journal of Inorganic Biochemistry, 102, 757-766.
http://dx.doi.org/10.1016/j.jinorgbio.2007.11.012
[18] Gomes, A.J., Espreafico, E.M. and Tfouni, E. (2013) Trans-[Ru(NO)Cl(Cyclam)](PF6)2 and [Ru(NO)(Hedta)] Incorporated in PLGA Nanoparticles for the Delivery of Nitric Oxide to B16-F10 Cells: Cytotoxicity and Phototoxicity. Molecular Pharmaceutics, 10, 3544-3554.
[19] Birnbaum, D., Kosmala, J. and Brannon-Peppas, L. (2000) Optimization of Preparation Techniques for Poly(Lactic Acid-Co-Glycolic Acid) Nanoparticles. Journal of Nanoparticle Research, 2, 173-181.
http://dx.doi.org/10.1023/A:1010038908767
[20] Kranz, H. and Bodmeier, R. (2007) A Novel in Situ Forming Drug Delivery System for Controlled Parenteral Drug Delivery. International Journal of Pharmaceutics, 332, 107-114.
http://dx.doi.org/10.1016/j.ijpharm.2006.09.033
[21] Vandervoort, J. and Ludwig, A. (2002) Biocompatible Stabilizers in the Preparation of PLGA Nanoparticles: A Factorial Design Study. International Journal of Pharmaceutics, 238, 77-92.
http://dx.doi.org/10.1016/S0378-5173(02)00058-3
[22] Wischke, C. and Schwendeman, S.P. (2008) Principles of Encapsulating Hydrophobic Drugs in PLA/PLGA Microparticles. International Journal of Pharmaceutics, 364, 298-327.
http://dx.doi.org/10.1016/j.ijpharm.2008.04.042
[23] Locatelli, E. and Comes Franchini, M. (2012) Biodegradable PLGA-b-PEG Polymeric Nanoparticles: Synthesis, Properties, and Nanomedical Applications as Drug Delivery System. Journal of Nanoparticle Research, 14, 1-17.
http://dx.doi.org/10.1007/s11051-012-1316-4
[24] Manchanda, R., Fernandez-Fernandez, A., Nagesetti, A. and McGoron, A.J. (2010) Preparation and Characterization of a Polymeric (PLGA) Nanoparticulate Drug Delivery System with Simultaneous Incorporation of Chemotherapeutic and Thermo-Optical Agents. Colloids and Surfaces B: Biointerfaces, 75, 260-267.
http://dx.doi.org/10.1016/j.colsurfb.2009.08.043
[25] Danhier, F., Ansorena, E., Silva, J.M., Coco, R., Le Breton, A. and Preat, V. (2012) PLGA-Based Nanoparticles: An Overview of Biomedical Applications. Journal of Controlled Release, 161, 505-522.
http://dx.doi.org/10.1016/j.jconrel.2012.01.043
[26] Makadia, H.K. and Siegel, S.J. (2011) Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier. Polymers (Basel), 3, 1377-1397.
http://dx.doi.org/10.3390/polym3031377
[27] Mao, S., Xu, J., Cai, C., Germershaus, O., Schaper, A. and Kissel, T. (2007) Effect of WOW Process Parameters on Morphology and Burst Release of FITC-Dextran Loaded PLGA Microspheres. International Journal of Pharmaceutics, 334, 137-148.
http://dx.doi.org/10.1016/j.ijpharm.2006.10.036
[28] Gomes, A.J., Assuncao, R.M.N., Rodrigues, G., Espreafico, E.M. and Machado, A.E.D. (2007) Preparation and Characterization of Poly(D,L-lactic-co-glycolic Acid) Nanoparticles Containing 3-(Benzoxazol-2-yl)-7-(N,N-diethyl ami- no)chromen-2-one. Journal of Applied Polymer Science, 105, 964-972.
http://dx.doi.org/10.1002/app.26204
[29] Gomes, A.J., Faustino, A.S., Lunardi, C.N., Lunardi, L.O. and Machado, A.E.H. (2007) Evaluation of Nanoparticles Loaded with Benzopsoralen in Rat Peritoneal Exudate Cells. International Journal of Pharmaceutics, 332, 153-160.
http://dx.doi.org/10.1016/j.ijpharm.2006.09.035
[30] Gomes, A.J., Lunardi, L.O., Marchetti, J.M., Lunardi, C.N. and Tedesco, A.C. (2005) Photobiological and Ultrastructural Studies of Nanoparticles of Poly(lactic-co-glycolic Acid)-Containing Bacteriochlorophyll-a as a Photosensitizer Useful for PDT Treatment. Drug Delivery, 12, 159-164.
http://dx.doi.org/10.1080/10717540590931846
[31] Gomes, A.J., Lunardi, C.N., Lunardi, L.O., Pitol, D.L. and Machado, A.E.H. (2008) Identification of Psoralen Loaded PLGA Microspheres in Rat Skin by Light Microscopy. Micron, 39, 40-44.
http://dx.doi.org/10.1016/j.micron.2007.06.015
[32] Gomes, A.J., Faustino, A.S., Machado, A.E.H., Zaniquelli, M.E.D., Rigoletto, T.D., Lunardi, C.N. and Lunardi, L.O. (2006) Characterization of PLGA Microparticles as a Drug Carrier for 3-Ethoxycarbonyl-2H-benzofuro[3,2-f]-1-ben- zopyran-2-one. Ultrastructural Study of Cellular Uptake and Intracellular Distribution. Drug Delivery, 13, 447-454.
http://dx.doi.org/10.1080/10717540600640369
[33] Tfouni, E., Truzzi, D.R., Tavares, A., Gomes, A.J., Figueiredo, L.E. and Franco, D.W. (2012) Biological Activity of Ruthenium Nitrosyl Complexes. Nitric Oxide-Biology and Chemistry, 26, 38-53.
http://dx.doi.org/10.1016/j.niox.2011.11.005
[34] Tfouni, E., Doro, F.G., Gomes, A.J., da Silva, R.S., Metzker, G., Benini, P.G.Z. and Franco, D.W. (2010) Immobilized Ruthenium Complexes and Aspects of Their Reactivity. Coordination Chemistry Reviews, 254, 355-371.
http://dx.doi.org/10.1016/j.ccr.2009.10.011
[35] Gomes, A.J., Lunardi, L.O., Caetano, F.H., Machado, A.E.H., Oliveira-Campos, A.M.F., Bendhack, L.M. and Lunardi, C.N. (2011) Biodegradable Nanoparticles Containing Benzopsoralens: An Attractive Strategy for Modifying Vascular Function in Pathological Skin Disorders. Journal of Applied Polymer Science, 121, 1348-1354.
http://dx.doi.org/10.1002/app.33427
[36] Mo, L., Hou, L., Guo, D., Xiao, X., Mao, P. and Yang, X. (2012) Preparation and Characterization of Teniposide PLGA Nanoparticles and Their Uptake in Human Glioblastoma U87MG Cells. International Journal of Pharmaceutics, 436, 815-824.
http://dx.doi.org/10.1016/j.ijpharm.2012.07.050
[37] Wang, Y.C., Xu, G.L., Jia, W.D., Han, S.J., Ren, W.H., Wang, W., Liu, W.B., Zhang, C.H. and Chen, H. (2012) Estrogen Suppresses Metastasis in Rat Hepatocellular Carcinoma through Decreasing Interleukin-6 and Hepatocyte Growth Factor Expression. Inflammation, 35, 143-149.
http://dx.doi.org/10.1007/s10753-011-9299-3
[38] Jyothi, N.V.N., Prasanna, P.M., Sakarkar, S.N., Prabha, K.S., Ramaiah, P.S. and Srawan, G.Y. (2010) Microencapsulation Techniques, Factors Influencing Encapsulation Efficiency. Journal of Microencapsulation, 27, 187-197.
http://dx.doi.org/10.3109/02652040903131301
[39] Song, H., Nie, S., Yang, X., Li, N., Xu, H., Zheng, L. and Pan, W. (2010) Characterization and in Vivo Evaluation of Novel Lipid-Chlorambucil Nanospheres Prepared Using a Mixture of Emulsifiers for Parenteral Administration. International Journal of Nanomedicine, 5, 933-942.
http://dx.doi.org/10.2147/IJN.S14596
[40] de Jesus Gomes, A., Lunardi, C.N., Caetano, F.H., Lunardi, L.O. and da Hora Machado, A.E. (2006) Phagocytosis of PLGA Microparticles in Rat Peritoneal Exudate Cells: A Time-Dependent Study. Microscopy and Microanalysis, 12, 399-405.
http://dx.doi.org/10.1017/S1431927606060284
[41] Gupta, H., Aqil, M., Khar, R.K., Ali, A., Bhatnagar, A. and Mittal, G. (2010) Sparfloxacin-Loaded PLGA Nanoparticles for Sustained Ocular Drug Delivery. Nanomedicine: Nanotechnology Biology and Medicine, 6, 324-333.
http://dx.doi.org/10.1016/j.nano.2009.10.004
[42] Gunasekaran, S., Kumaresan, S., Balaji, R.A., Anand, G. and Seshadri, S. (2008) Vibrational Spectra and Normal Coordinate Analysis on Structure of Chlorambucil and Thioguanine. Pramana: Journal of Physics, 71, 1291-1300.
http://dx.doi.org/10.1007/s12043-008-0183-0
[43] Yang, J., Lee, C.H., Park, J., Seo, S., Lim, E.K., Song, Y.J., Suh, J.S., Yoon, H.G., Huh, Y.M. and Haam, S. (2007) Antibody Conjugated Magnetic PLGA Nanoparticles for Diagnosis and Treatment of Breast Cancer. Journal of Materials Chemistry, 17, 2695-2699.
http://dx.doi.org/10.1039/b702538f
[44] Garcia, X., Escribano, E., Domenech, J., Queralt, J. and Freixes, J. (2011) In Vitro Characterization and in Vivo Analgesic and Anti-Allodynic Activity of PLGA-Bupivacaine Nanoparticles. Journal of Nanoparticle Research, 13, 2213- 2223.
http://dx.doi.org/10.1007/s11051-010-9979-1
[45] Mi, F.L., Lin, Y.M., Wu, Y.B., Shyu, S.S. and Tsai, Y.H. (2002) Chitin/PLGA Blend Microspheres as a Biodegradable Drug-Delivery System: Phase-Separation, Degradation and Release Behavior. Biomaterials, 23, 3257-3267.
http://dx.doi.org/10.1016/S0142-9612(02)00084-4
[46] Namazi, H. and Jafarirad, S. (2011) In Vitro Photo-Controlled Drug Release System Based on Amphiphilic Linear-Dendritic Diblock Copolymers; Self-Assembly Behavior and Application as Nanocarrier. Journal of Pharmaceutical Sciences, 14, 162-180.
[47] Ray, R.S., Rana, B., Swami, B., Venu, V. and Chatterjee, M. (2006) Vanadium Mediated Apoptosis and Cell Cycle Arrest in MCF7 Cell Line. Chemico-Biological Interactions, 163, 239-247.
http://dx.doi.org/10.1016/j.cbi.2006.08.006
[48] Fonseca, C., Simoes, S. and Gaspar, R. (2002) Paclitaxel-Loaded PLGA Nanoparticles: Preparation, Physicochemical Characterization and in Vitro Anti-Tumoral Activity. Journal of Controlled Release, 83, 273-286.
http://dx.doi.org/10.1016/S0168-3659(02)00212-2
[49] Averineni, R.K.S., Shavi, G.V., Gurram, A.K., Deshpande, P.B., Arumugam, K., Maliyakkal, N., Meka, S.R. and Nayahirama, U. (2012) PLGA 50:50 Nanoparticles of Paclitaxel: Development, in Vitro Anti-Tumor Activity in BT-549 Cells and in Vivo Evaluation. Bulletin of Materials Science, 35, 319-326.
http://dx.doi.org/10.1007/s12034-012-0313-7
[50] Mukerjee, A. and Vishwanatha, J.K. (2009) Formulation, Characterization and Evaluation of Curcumin-Loaded PLGA Nanospheres for Cancer Therapy. Anticancer Research, 29, 3867-3875.
[51] Pirooznia, N., Hasannia, S., Lotfi, A.S. and Ghanei, M. (2012) Encapsulation of Alpha-1 Antitrypsin in PLGA Nanoparticles: In Vitro Characterization as an Effective Aerosol Formulation in Pulmonary Diseases. Journal of Nanobiotechnology, 10, 1-15.
[52] Ribeiro-Costa, R.M., Alves, A.J., Santos, N.P., Nascimento, S.C., Goncalves, E.C., Silva, N.H., Honda, N.K. and Santos-Magalhaes, N.S. (2004) In Vitro and in Vivo Properties of Usnic Acid Encapsulated into PLGA-Microspheres. Journal of Microencapsulation, 21, 371-384.
http://dx.doi.org/10.1080/02652040410001673919                                                   eww150109lx

Colorectal Cancer Surgery in Extreme Elderly Population

Read  full  paper  at:

http://www.scirp.org/journal/PaperInformation.aspx?PaperID=52682#.VKC8gcCAM4

Author(s)

Javier Gallego Plazas1*, Elena Asensio1, Juan C. Navalon2, Inmaculada Lozano1, Jose M. Navarro2, Guillermo Ricote1, Montse Olcina2, Almudena Cotes1, Henry M. Ore1, Alejandra Magdaleno1, Maria C. Ors1, Maria J. Escudero1, Miguel A. Morcillo2

Affiliation(s)

1Servicio de Oncología Médica, Hospital General Universitario de Elche-Hospital Vega Baja, Alicante, Espana.
2Servicio de Cirugía General, Hospital Vega Baja, Alicante, Espana.

ABSTRACT

Background: Colorectal cancer surgery in extreme elderly population (380 years) is a growing problem and a subject in question. Experiences and results in this scenario are limited. Methods: Medical records of patients 380 years old hospitalized in General Surgery Department in Vega Baja Hospital between January 1st 2008 and December 31st 2012 were retrospectively reviewed. Patients diagnosed with colorectal cancer (CRC) were selected. Data regarding comorbidities, treatment performed, complications and survival were collected. Results: 95 patients (74 colon, 21 rectum) were identified: median age 83.0 years (80 – 93), 64.2% males. 93.7% of patients were operated on: median hospital and post-operative stay of 11 and 9 days, respectively; curative intention surgery 82.1%. 4.2% of patients were treated with interventionist procedures, and 2.1% of patients in a conservative way. 17.9% and 4.2% of patients came up with early and late complications, respectively. Early and late mortality occurred in 9.5% and 3.2% of patients, respectively. By March 2014, with a median follow up of 43.8 months, median overall survival for colorectal cancer patients was 2.7 years (95%IC, 2.0 – 3.2). Univariaye Cox Regresion analysis revealed the presence of cardiomyopathy (p = 0.024), the presence of chornic kidney disease (p = 0.025), the presence of comorbidities (vs absence) (0.026), the number of comorbidities (0.034), type of admission (p = 0.001), treatment with surgery (p = 0.001) and the incidence of early (p = 0.004) or late complications (p = 0.023) associated to overall survival with statically significance. Multivariate Cox Regression analysis showed number of comorbidities (HR = 1.104; 95%CI: 0.851 – 1.431; p = 0.456), treatment with surgery (HR = 4.928; 95%CI: 1.815 – 13.385; p = 0.002), programmed admission into hospital (HR = 2.316; 95%CI: 1.298 – 4.133; p = 0.004), and the incidence of late complications (HR = 4.629; 95%CI: 1.279 – 16.750; p = 0.020) independently associated with overall survival. Interaction test between number of comorbidities and early complication was performed (HR = 1.453; 95%CI: 0.971 – 2.175; p = 0.070). Conclusions: In our experience surgery for CRC patients may increase overall survival even in an extreme elderly population (380 years). Nevertheless when considering surgery for CRC in this subgroup of patients, factor such as type of admission into hospital and comorbidities should be taken into account in order to optimize treatment results in the effort to individualize CRC management in this growing population.

KEYWORDS

Colorectal, Cancer, Surgery, Elderly

Cite this paper

Plazas, J. , Asensio, E. , Navalon, J. , Lozano, I. , Navarro, J. , Ricote, G. , Olcina, M. , Cotes, A. , Ore, H. , Magdaleno, A. , Ors, M. , Escudero, M. and Morcillo, M. (2015) Colorectal Cancer Surgery in Extreme Elderly Population. Journal of Cancer Therapy, 6, 12-20. doi: 10.4236/jct.2015.61002.

References

[1] van de Velde, C.J.H., Boelens, P.G., Tanis, P.J., et al. (2014) Experts Reviews of the Multidisciplinary Consensus Conference Colon and Rectal Cancer 2012: Science, Opinions and Experiences from the Experts of Surgery. European Journal of Surgical Oncology, 40, 454-468.
http://dx.doi.org/10.1016/j.ejso.2013.10.013
[2] http://www.ncin.org.uk/cancer_information_tools/ukcis
[3] Brenner, H., Bouvier, A.M., Foschi, R., et al. (2012) Progress in Colorectal Cancer Survival in Europe from the Late 1980s to the Early 21st Century: The EUROCARE Study. International Journal of Cancer, 131, 1649-1658. http://dx.doi.org/10.1002/ijc.26192
[4] Jensen, H.E., Nielsen, J. and Balslev, I. (1970) Carcinoma of the Colon in Old Age. Annals of Surgery, 171, 107-115. http://dx.doi.org/10.1097/00000658-197001000-00016
[5] Irvin, T.T. (1988) Prognosis of Colorectal Cancer in the Elderly. British Journal of Surgery, 75, 419-421.
[6] Boyd, J.B., Bradford Jr., B. and Watne, A.L. (1980) Operative Risk Factors of Colon Resection in the Elderly. Annals of Surgery, 192, 743-746. http://dx.doi.org/10.1097/00000658-198012000-00009
[7] Dekker, J.W., Gooiker, G.A., Bastiaannet, E., et al. (2014) Cause of Death the First Year after Curative Colorectal Cancer Surgery; a Prolonged Impact of the Surgery in Elderly Colorectal Cancer Patients. European Journal of Surgical Oncology, 40, 1481-1487. http://dx.doi.org/10.1016/j.ejso.2014.05.010
[8] Kolfschoten, N.E., Wouters, M.W., Gooiker, G.A., et al. (2012) Nonelective Colon Cancer Resections in Elderly Patients: Results from the Dutch Surgical Colorectal Audit. Digestive Surgery, 29, 412-419.
http://dx.doi.org/10.1159/000345614
[9] Mamidanna, R., Eid-Arimoku, L., Almoudaris, A.M., et al. (2012) Poor 1-Year Survival in Elderly Patients Undergoing Nonelective Colorectal Resection. Diseases of the Colon & Rectum, 55, 788-796.
http://dx.doi.org/10.1097/DCR.0b013e3182585a35
[10] Jemal, A., Murray, T., Ward, E., et al. (2005) Cancer Statistics, 2005. CA: A Cancer Journal for Clinicians, 55, 10-30. http://dx.doi.org/10.3322/canjclin.55.1.10
[11] Goldberg, R.M., Tabah-Fisch, I., Bleiberg, H., de Gramont, A., Tournigand, C., Andre, T., et al. (2006) Pooled Analysis of Safety and Efficacy of Oxaliplatin plus Fluorouracil/Leucovorin Administered Bimonthly in Elderly Patients with Colorectal Cancer. Journal of Clinical Oncology, 24, 4085-4091.
http://dx.doi.org/10.1200/JCO.2006.06.9039
[12] Ries, L.A.G., Harkins, D., Krapcho, M., et al., Eds. (2003) Contents of the SEER Cancer Statistics Review, 1975-2003. National Cancer Institute, Bethesda. http://seer.cancer.gov/csr/1975_2003/
[13] Edwards, B.K., Howe, H.L., Ries, L.A.G., Thun, M.J., Rosenberg, H.M., Yancik, R., et al. (2002) Annual Report to the Nation on the Status of Cancer, 1973-1999, Featuring Implications of Age and Aging on US Cancer Burden. Cancer, 94, 2766-2792. http://dx.doi.org/10.1002/cncr.10593
[14] Minino, A.M., Heron, M.P., Murphy, S.L., et al. (2007) Deaths: Final Data for 2004. National Vital Statistics Reports, 55, 1-119.
[15] (2007) Life Expectancy by Selected Ages since 1946: Females. Institut National D’tudes Démographiques, Paris. http://www.ined.fr/fichier/t_telechargement/11151/telechargement_fichier_fr_telechargement_fichier_ fr_sd2004_t69esp_fm.1.xls
[16] (2007) Life Expectancy by Selected Ages since 1946: Males. Institut National D’tudes Démographiques, Paris. http://www.ined.fr/fichier/t_telechargement/11152/telechargement_fichier_fr_telechargement_fichier_ fr_sd2004_t69esp_fm.1.xls
[17] Mitry, E., Bouvier, A.M., Esteve, J. and Faivre, J. (2005) Improvement in Colorectal Cancer Survival: A Population-Based Study. European Journal of Cancer, 41, 2297-2303.
http://dx.doi.org/10.1016/j.ejca.2005.01.028
[18] Jemal, A., Siegel, R., Ward, E., Hao, Y., Xu, J., Murray, T. and Thun, M.J. (2008) Cancer Statistics, 2008. CA: A Cancer Journal for Clinicians, 58, 71-96. http://dx.doi.org/10.3322/CA.2007.0010
[19] Yancik, R., Havlik, R.J., Wesley, M.N., Ries, L., Long, S., Rossi, W.K. and Edwards, B.K. (1996) Cancer and Comorbidity in Older Patients: A Descriptive Profile. Annals of Epidemiology, 6, 399-412.
http://dx.doi.org/10.1016/S1047-2797(96)00063-4
[20] Coebergh, J.W.W., Janssen-Heijnen, M.L.G., Post, P.N. and Razenberg, P.P.A. (1999) Serious Comorbidity among Unselected Cancer Patients Newly Diagnosed in the Southeastern Part of The Netherlands in 1993-1996. Journal of Clinical Epidemiology, 52, 1131-1136.
http://dx.doi.org/10.1016/S0895-4356(99)00098-0
[21] Zeber, J.E., Copeland, L.A., Hosek, B.J., Karnad, A.B., Lawrence, V.A. and Sanchez-Reilly, S.E. (2008) Cancer Rates, Medical Comorbidities, and Treatment Modalities in the Oldest Patients. Critical Reviews in Oncology/Hematology, 67, 237-242. http://dx.doi.org/10.1016/j.critrevonc.2008.02.002
[22] Piccirillo, J.F., Tierney, R.M., Costas, I., Grove, L. and Spitznagel Jr., E.L. (2004) Prognostic Importance of Comorbidity in a Hospitalbased Cancer Registry. Journal of the American Medical Association, 291, 2441-2447. http://dx.doi.org/10.1001/jama.291.20.2441
[23] Geraci, J.M., Escalante, C.P., Freeman, J.L. and Goodwin, J.S. (2005) Comorbid Disease and Cancer: The Need for More Relevant Conceptual Models in Health Services Research. Journal of Clinical Oncology, 23, 7399-7404. http://dx.doi.org/10.1200/JCO.2004.00.9753
[24] Koroukian, S.M., Murray, P. and Madigan, E. (2006) Comorbidity, Disability, and Geriatric Syndromes in Elderly Cancer Patients Receiving Home Health Care. Journal of Clinical Oncology, 24, 2304-2310.
http://dx.doi.org/10.1200/JCO.2005.03.1567
[25] Extermann, M., Overcash, J., Lyman, G.H., Parr, J. and Balducci, L. (1998) Comorbidity and Functional Status Are Independent in Older Cancer Patients. Journal of Clinical Oncology, 16, 1582-1587.
[26] Read, W.L., Tierney, R.M., Page, N.C., Costas, I., Govindan, R., Spitznagel, E.L.J. and Piccirillo, J.F. (2004) Differential Prognostic Impact of Comorbidity. Journal of Clinical Oncology, 22, 3099-3103.
http://dx.doi.org/10.1200/JCO.2004.08.040
[27] Gooiker, G.A., Dekker, J.W.T., Bastiaannet, E., van der Geest, L.G.M., Merkus, J.W.S., van de Velde, C.J.H., et al. (2012) Risk Factors for Excess Mortality in the First Year after Curative Surgery for Colorectal Cancer. Annals of Surgical Oncology, 19, 2428-2434. http://dx.doi.org/10.1245/s10434-012-2294-6                   eww141229lx

The Mitochondrial Pyruvate Carrier and Metabolic Regulation

Read  full  paper  at:

http://www.scirp.org/journal/PaperInformation.aspx?PaperID=52150#.VIZK0WfHRK0

Author(s)

ABSTRACT

Pyruvate is a key intermediate at the branchpoint of anaerobic and aerobic energy metabolism. Its transport into the mitochondrial matrix is necessary prior to its decarboxylation into acetyl-CoA, which feeds the reducing equivalent-generating tricarboxylic acid (TCA) cycle. Although the existence of specific carrier transport of cytosolic pyruvate into the mitochondria has been inferred from a myriad of studies, the identities of the mitochondrial pyruvate carrier (MPC) were only confirmed very recently. Identification of the MPC facilitated several other recent advances. These include the finding of MPC’s inhibition by the insulin-sensitizing drug family thiazolidinediones, how cells respond flexibly to a reduction in MPC functionality, as well as insights into how changes in MPC levels affect oncogenic potential of cancer cells. These new findings, discussed here in this brief review, have important implications in therapeutic approaches towards metabolic disorders and cancer.

Cite this paper

Tang, B. (2014) The Mitochondrial Pyruvate Carrier and Metabolic Regulation. CellBio, 3, 111-117. doi: 10.4236/cellbio.2014.34011.

References

[1] Papa, S., Francavilla, A., Paradies, G. and Meduri, B. (1971) The Transport of Pyruvate in Rat Liver Mitochondria. FEBS Letters, 12, 285-288.
http://dx.doi.org/10.1016/0014-5793(71)80200-4
[2] Halestrap, A.P. and Denton, R.M. (1974) Specific Inhibition of Pyruvate Transport in Rat Liver Mitochondria and Human Erythrocytes by alpha-Cyano-4-hydroxycinnamate. Biochemical Journal, 138, 313-316.
[3] Halestrap, A.P. and Denton, R.M. (1975) The Specificity and Metabolic Implications of the Inhibition of Pyruvate Transport in Isolated Mitochondria and Intact Tissue Preparations by alpha-Cyano-4-hydroxycinnamate and Related Compounds. Biochemical Journal, 148, 97-106.
[4] Thomas, A.P. and Halestrap, A.P. (1981) Identification of the Protein Responsible for Pyruvate Transport into Rat Liver and Heart Mitochondria by Specific Labelling with [3H]N-Phenylmaleimide. Biochemical Journal, 196, 471-479.
[5] Schell, J.C. and Rutter, J. (2013) The Long and Winding Road to the Mitochondrial Pyruvate Carrier. Cancer & Metabolism, 1, 6.
[6] Gray, L.R., Tompkins, S.C. and Taylor, E.B. (2014) Regulation of Pyruvate Metabolism and Human Disease. Cellular and Molecular Life Sciences, 71, 2577-2604.
http://dx.doi.org/10.1007/s00018-013-1539-2
[7] Jeoung, N.H., Harris, C.R. and Harris, R.A. (2014) Regulation of Pyruvate Metabolism in Metabolic-Related Diseases. Reviews in Endocrine & Metabolic Disorders, 15, 99-110.
http://dx.doi.org/10.1007/s11154-013-9284-2
[8] Palsson-McDermott, E.M. and O’Neill, L.A.J. (2013) The Warburg Effect Then and Now: From Cancer to Inflammatory Diseases. Bioessays, 35, 965-973.
http://dx.doi.org/10.1002/bies.201300084
[9] Warburg, O., Wind, F. and Negelein, E. (1927) The Metabolism of Tumors in the Body. The Journal of General Physiology, 8, 519-530.
http://dx.doi.org/10.1085/jgp.8.6.519
[10] Iqbal, M.A., Gupta, V., Gopinath, P., Mazurek, S. and Bamezai, R.N.K. (2014) Pyruvate Kinase M2 and Cancer: An Updated Assessment. FEBS Letters, 588, 2685-2692.
http://dx.doi.org/10.1016/j.febslet.2014.04.011
[11] Pande, S.V. and Parvin, R. (1978) Pyruvate and Acetoacetate Transport in Mitochondria. A Reappraisal. Journal of Biological Chemistry, 253, 1565-1573.
[12] Shearman, M.S. and Halestrap, A.P. (1984) The Concentration of the Mitochondrial Pyruvate Carrier in Rat Liver and Heart Mitochondria Determined with Alpha-Cyano-Beta-(1-phenylindol-3-yl)acrylate. Biochemical Journal, 223, 673- 676.
[13] Bricker, D.K., Taylor, E.B., Schell, J.C., Orsak, T., Boutron, A., Chen, Y.C., Cox, J.E., Cardon, C.M., Van Vranken, J.G., Dephoure, N., Redin, C., Boudina, S., Gygi, S.P., Brivet, M., Thummel, C.S. and Rutter, J. (2012) A Mitochondrial Pyruvate Carrier Required for Pyruvate Uptake in Yeast, Drosophila, and Humans. Science, 337, 96-100.
http://dx.doi.org/10.1126/science.1218099
[14] Herzig, S., Raemy, E., Montessuit, S., Veuthey, J.L., Zamboni, N., Westermann, B., Kunji, E.R.S. and Martinou, J.C. (2012) Identification and Functional Expression of the Mitochondrial Pyruvate Carrier. Science, 337, 93-96.
http://dx.doi.org/10.1126/science.1218530
[15] Brailsford, M.A., Thompson, A.G., Kaderbhai, N. and Beechey, R.B. (1986) The Extraction and Reconstitution of the Alpha-Cyanocinnamate-Sensitive Pyruvate Transporter from Castor Bean Mitochondria. Biochemical and Biophysical Research Communications, 140, 1036-1042.
http://dx.doi.org/10.1016/0006-291X(86)90739-4
[16] Nalecz, M.J., Nalecz, K.A., Broger, C., Bolli, R., Wojtczak, L. and Azzi, A. (1986) Extraction, Partial Purification and Functional Reconstitution of Two Mitochondrial Carriers Transporting Keto Acids: 2-Oxoglutarate and Pyruvate. FEBS Letters, 196, 331-336.
http://dx.doi.org/10.1016/0014-5793(86)80273-3
[17] Capuano, F., Di Paola, M., Azzi, A. and Papa, S. (1990) The Monocarboxylate Carrier from Rat Liver Mitochondria. Purification and Kinetic Characterization in a Reconstituted System. FEBS Letters, 261, 39-42.
http://dx.doi.org/10.1016/0014-5793(90)80631-R
[18] Nalecz, M.J., Nalecz, K.A. and Azzi, A. (1991) Purification and Functional Characterisation of the Pyruvate (Monocarboxylate) Carrier from Baker’s Yeast Mitochondria (Saccharomyces cerevisiae). Biochimica et Biophysica Acta, 1079, 87-95.
http://dx.doi.org/10.1016/0167-4838(91)90028-X
[19] Hildyard, J.C.W. and Halestrap, A.P. (2003) Identification of the Mitochondrial Pyruvate Carrier in Saccharomyces cerevisiae. Biochemical Journal, 374, 607-611.
http://dx.doi.org/10.1042/BJ20030995
[20] Todisco, S., Agrimi, G., Castegna, A. and Palmieri, F. (2006) Identification of the Mitochondrial NAD+ Transporter in Saccharomyces cerevisiae. Journal of Biological Chemistry, 281, 1524-1531.
http://dx.doi.org/10.1074/jbc.M510425200
[21] Da Cruz, S., Xenarios, I., Langridge, J., Vilbois, F., Parone, P.A. and Martinou, J.C. (2003) Proteomic Analysis of the Mouse Liver Mitochondrial Inner Membrane. Journal of Biological Chemistry, 278, 41566-41571.
http://dx.doi.org/10.1074/jbc.M304940200
[22] Halestrap, A.P. (2012) The Mitochondrial Pyruvate Carrier: Has It Been Unearthed at Last? Cell Metabolism, 16, 141-143.
http://dx.doi.org/10.1016/j.cmet.2012.07.013
[23] Li, C.L., Wang, M., Ma, X.Y. and Zhang, W. (2014) NRGA1, a Putative Mitochondrial Pyruvate Carrier, Mediates ABA Regulation of Guard Cell Ion Channels and Drought Stress Responses in Arabidopsis. Molecular Plant, 7, 1508-1521.
http://dx.doi.org/10.1093/mp/ssu061
[24] Lehmann, J.M., Moore, L.B., Smith-Oliver, T.A., Wilkison, W.O., Willson, T.M. and Kliewer, S.A. (1995) An Antidiabetic Thiazolidinedione Is a High Affinity Ligand for Peroxisome Proliferator-Activated Receptor γ (PPARγ). Journal of Biological Chemistry, 270, 12953-12956.
http://dx.doi.org/10.1074/jbc.270.22.12953
[25] Divakaruni, A.S., Wiley, S.E., Rogers, G.W., Andreyev, A.Y., Petrosyan, S., Loviscach, M., Wall, E.A., Yadava, N., Heuck, A.P., Ferrick, D.A., Henry, R.R., McDonald, W.G., Colca, J.R., Simon, M.I., Ciaraldi, T.P. and Murphy, A.N. (2013) Thiazolidinediones Are Acute, Specific Inhibitors of the Mitochondrial Pyruvate Carrier. Proceedings of the National Academy of Sciences of the United States of America, 110, 5422-5427.
http://dx.doi.org/10.1073/pnas.1303360110
[26] Hardie, D.G. (2014) AMPK: Positive and Negative Regulation, and Its Role in Whole-Body Energy Homeostasis. Current Opinion in Cell Biology, 33C, 1-7.
[27] Colca, J.R., McDonald, W.G., Cavey, G.S., Cole, S.L., Holewa, D.D., Brightwell-Conrad, A.S., Wolfe, C.L., Wheeler, J.S., Coulter, K.R., Kilkuskie, P.M., Gracheva, E., Korshunova, Y., Trusgnich, M., Karr, R., Wiley, S.E., Divakaruni, A.S., Murphy, A.N., Vigueira, P.A., Finck, B.N. and Kletzien, R.F. (2013) Identification of a Mitochondrial Target of Thiazolidinedione Insulin Sensitizers (mTOT)—Relationship to Newly Identified Mitochondrial Pyruvate Carrier Proteins. PLoS ONE, 8, e61551.
http://dx.doi.org/10.1371/journal.pone.0061551
[28] Vigueira, P.A., McCommis, K.S., Schweitzer, G.G., Remedi, M.S., Chambers, K.T., Fu, X., McDonald, W.G., Cole, S.L., Colca, J.R., Kletzien, R.F., Burgess, S.C. and Finck, B.N. (2014) Mitochondrial Pyruvate Carrier 2 Hypomorphism in Mice Leads to Defects in Glucose-Stimulated Insulin Secretion. Cell Reports, 7, 2042-2053.
http://dx.doi.org/10.1016/j.celrep.2014.05.017
[29] Vacanti, N.M., Divakaruni, A.S., Green, C.R., Parker, S.J., Henry, R.R., Ciaraldi, T.P., Murphy, A.N. and Metallo, C.M. (2014) Regulation of Substrate Utilization by the Mitochondrial Pyruvate Carrier. Molecular Cell, 56, 425-435.
http://dx.doi.org/10.1016/j.molcel.2014.09.024
[30] Yang, C., Sudderth, J., Dang, T., Bachoo, R.M., Bachoo, R.G., McDonald, J.G. and DeBerardinis, R.J. (2009) Glioblastoma Cells Require Glutamate Dehydrogenase to Survive Impairments of Glucose Metabolism or Akt Signaling. Cancer Research, 69, 7986-7993.
http://dx.doi.org/10.1158/0008-5472.CAN-09-2266
[31] Yang, C., Ko, B., Hensley, C.T., Jiang, L., Wasti, A.T., Kim, J., Sudderth, J., Calvaruso, M.A., Lumata, L., Mitsche, M., Rutter, J., Merritt, M.E. and DeBerardinis, R.J. (2014) Glutamine Oxidation Maintains the TCA Cycle and Cell Survival during Impaired Mitochondrial Pyruvate Transport. Molecular Cell, 56, 414-424.
http://dx.doi.org/10.1016/j.molcel.2014.09.025
[32] Cairns, R.A., Harris, I.S. and Mak, T.W. (2011) Regulation of Cancer Cell Metabolism. Nature Reviews Cancer, 11, 85-95.
http://dx.doi.org/10.1038/nrc2981
[33] Schell, J.C., Olson, K.A., Jiang, L., Hawkins, A.J., Van Vranken, J.G., Xie, J., Egnatchik, R.A., Earl, E.G., DeBerardinis, R.J. and Rutter, J. (2014) A Role for the Mitochondrial Pyruvate Carrier as a Repressor of the Warburg Effect and Colon Cancer Cell Growth. Molecular Cell, 56, 400-413.
http://dx.doi.org/10.1016/j.molcel.2014.09.026
[34] Moreb, J.S. (2008) Aldehyde Dehydrogenase as a Marker for Stem Cells. Current Stem Cell Research & Therapy, 3, 237-246.
http://dx.doi.org/10.2174/157488808786734006
[35] Tirino, V., Desiderio, V., Paino, F., De Rosa, A., Papaccio, F., La Noce, M., Laino, L., De Francesco, F. and Papaccio, G. (2013) Cancer Stem Cells in Solid Tumors: An Overview and New Approaches for Their Isolation and Characterization. FASEB Journal, 27, 13-24.
http://dx.doi.org/10.1096/fj.12-218222
[36] Leushacke, M. and Barker, N. (2012) Lgr5 and Lgr6 as Markers to Study Adult Stem Cell Roles in Self-Renewal and Cancer. Oncogene, 31, 3009-3022.
http://dx.doi.org/10.1038/onc.2011.479
[37] Zhou, J., Ng, S.B. and Chng, W.J. (2013) LIN28/LIN28B: An Emerging Oncogenic Driver in Cancer Stem Cells. International Journal of Biochemistry & Cell Biology, 45, 973-978.
http://dx.doi.org/10.1016/j.biocel.2013.02.006
[38] Iv Santaliz-Ruiz, L.E., Xie, X., Old, M., Teknos, T.N. and Pan, Q. (2014) Emerging Role of Nanog in Tumorigenesis and Cancer Stem Cells. International Journal of Cancer, 135, 2741-2748.
http://dx.doi.org/10.1002/ijc.28690                                                                                               eww141209lx

Epidemiological and Histological Aspects of Women Genital Cancers in Côte d’Ivoire

Read  full  paper  at:

http://www.scirp.org/journal/PaperInformation.aspx?PaperID=47155#.VGAnI2fHRK0

Author(s)

Kouamé Justin N’Dah, Brahima Doukoure, Emile Troh, N’Guessan Alphonse Aman, Kouakou Emmanuel Koffi, Arthur Didier Kouamé, Ahoua Benjamin Effi, Ahoua François de Salès D’Horpock, Mohénou Isidore Jean-Marie Diomande

Affiliation(s)

Laboratory of Pathology, University Hospital of Cocody, Abidjan, Cote d’Ivoire.
Laboratory of Pathology, University Hospital of Cocody, Abidjan, Cote d’Ivoire.
Laboratory of Pathology, University Hospital of Cocody, Abidjan, Cote d’Ivoire.
Laboratory of Pathology, University Hospital of Bouake, Bouake, Cote d’Ivoire.
Laboratory of Pathology, University Hospital of Treichville, Abidjan, Cote d’Ivoire.
Department of Obstetrical Gynecology, University Hospital of Cocody, Abidjan, Cote d’Ivoire.
Laboratory of Pathology, University Hospital of Bouake, Bouake, Cote d’Ivoire.
Laboratory of Pathology, University Hospital of Treichville, Abidjan, Cote d’Ivoire.
Laboratory of Pathology, University Hospital of Cocody, Abidjan, Cote d’Ivoire.

ABSTRACT

Genital cancers in women play an important role in cancer pathology in the developing countries. Objectives: This study aimed at clarifying the epidemiological and histopathological aspects of woman genital cancers in C?te d’Ivoire. Materials and Methods: This was a retrospective and descriptive study conducted in the anatomical pathology laboratories of the university hospitals in Abidjan. The study was carried out over a period of twenty-four years (1984-2007). The variables studied were: frequency, age, histological type and prognosis. Results: Genital cancers in women accounted for 41.28% (n = 2491) of cancer in women and 21% of cancers. The average age of patients was 46.65 years (range 2 – 88 years). The cervix (82.85%) was the main location followed by the ovary (8.6%). Histologically, carcinomas (92.93%) were the most common types of sarcomas. Cervical cancers were diagnosed at an average age of 47.36 years (range 2 to 88 years). Carcinomas were the predominant histological type (92.88%) with 57.4% (n = 450) diagnosed at pT2N0M0 stage. The average age of patients bearing ovarian cancer was 39.13 years (range 8 to 82 years).Common malignant epithelial tumours were the most common histological type (57.48%) (n = 123) followed by non-Hodgkin lymphoma (14.95%). Conclusion: Genital cancers in women are common and poorly prognosed in Cote d’Ivoire. The adoption of a policy of routine screening is needed to improve the prognosis of these tumours.

KEYWORDS

Cancer, Genital, Women, Epidemiology-Histology-Côte d’Ivoire

Cite this paper

N’Dah, K. , Doukoure, B. , Troh, E. , Aman, N. , Koffi, K. , Kouamé, A. , Effi, A. , D’Horpock, A. and Diomande, M. (2014) Epidemiological and Histological Aspects of Women Genital Cancers in Côte d’Ivoire. Open Journal of Obstetrics and Gynecology, 4, 516-523. doi: 10.4236/ojog.2014.49073.

References

[1] Chbani, L., Hafid, I., Berraho, M., et al. (2013) Epidemiological and Pathological Aspects of Cancers in the Regions of Fez-Boulemane (Morocco). Eastern Mediterranean Health Journal, 19, 264-270.
[2] International Agency for Research on Cancer, Globocancer (2008) Cancer Incidence, Mortality, and Prevalence Worldwide in 2008. http://globobcancer.iarc.fr/
[3] Amegbor, K., Darre, T., Ayena, K.D., et al. (2011) Cancers in Togo from 1984 to 2008. Epidemiological and Pathological Aspects of 5251 Cases. Journal of Cancer Epidemiology, 3, 98-102.
[4] N’Dahindwa, V., Ngendahayo, L. and Vyankandondera, J. (2012) Epidemiological and Pathological Aspects of Cancer in Hospital University Teaching of Rwanda. Medical Journal of Rwanda, 69, 40-48.
[5] Goumbri/Lompo, O.M., Domagni, O.E., Sanopu, A.M., et al. (2009) Epidemiological and Histopathological Aspects of Cancers in Burkina Faso. African Journal of Cancer, 1, 207-211.
[6] Effi, A.B., N’Dah, K.J., N’Guiessan, A.A., et al. (2012) Epidemiology and Pathology of Cancers in Ivory Coast. African Journal of Cancer, 4, 41-47.
[7] Diop, P.S., Ka, I., N’Diaye, N., et al. (2012) Breast and Gynecological Cancers in the General Hospital of Grand Yoff Dakar: Analysis and Implications for Epidemiological Aspects about 1269 Cases. African Journal of Cancer, 4, 176-179.
[8] Platz, C.E. and Benda, J.A. (1995) Female Genital Tract Cancers. Cancer, 75, 270-294. http://dx.doi.org/10.1002/1097-0142(19950101)75:1+<270::AID-CNCR2820751312>3.0.CO;2-D
[9] Traore, M., Diabate, F.S., Diarra, I., et al. (2004) Breast and Gynecological Cancers: Epidemiological and Clinical Aspects of Hospital Point G in Bamako. Medical Journal of Mali, 19, 4-9.
[10] Mouele Sone, A., Essomba, M., Fezeu, M., et al. (1994) Importance of Radiotherapy in the Treatment of Cervical Cancer in Developing Countries. Journal of African Medicine, 41, 187.
[11] Banza, K., Kizonde, J., Unga, M., et al. (1999) Cervical Cancer: Issues of Management. About 40 Cases Seen at the hospital in Lubumbashi. Journal of Lumumbashi, 2, 17-20.
[12] Abboud, J., Attieh, E. and Germanos, A. (1992) Invasive Cervical Cancer. Treatment Tailored to the Therapeutic Method. Journal of Obstetrics & Gynaecology, 21, 19-22.
[13] Sankaranarayanan, R., Blanck, R.J., Swaninatha, R., et al. (1998) An Overview of Cancer Survival in Developing Countries. IARC Scientific Publications, 145, 135-173.
[14] Querleu, D. and White, E. (2005) Cervical Cancer. Treaty Gynecology, Medicine-Sciences, Flammarion Editions, Paris, 359-360.
[15] Nayama, M., Nouhou, H., Madougou-Sunna, K., et al. (2006) Breast and Gynecological Cancers: Epidemiological and Histological Aspects Made from the Record of Pathology Laboratory of the Faculty of Science and Heath Niamey (Niger). Medical Journal of Mali, 31, 43-49.
[16] Mandelblatt, J.S., Kanetsky, P., Eggert, L. and Gold, K. (1999) Is HIV Infection a Cofactor for Cervical Squamous Cell Neoplasia? Cancer Epidemiology, Biomarkers & Prevention, 8, 97-106.
[17] Uzoiwe, S.A. and Seleye-Fubara, D. (2004) Cervical Cancers in Fort Harcourt, Rivers State. A Clinico-Pathological 13 Years Review. Nigerian Journal of Medicine, 13, 110-113.
[18] Sankaranarayanan, R. and Ferley, J. (2006) Worldwide Burden of Gynecological Cancer: The Size of the Problem. Best Practice & Research Clinical Obstetrics & Gynaecology, 20, 207-225.
http://dx.doi.org/10.1016/j.bpobgyn.2005.10.007
[19] Statistics about Ovarian Cancers (2013) http://www.cureresearch.com/o/Ovarian_Cancer/stat-country.htm
[20] Duarte-Franco, E. and Franco, E.L. (2004) Other Gynecologic Cancers: Endometrial, Ovarian, Vulvar and Vaginal Cancers. BMC Women’s Health, 4, S14. http://dx.doi.org/10.1186/1472-6874-4-S1-S14
[21] Body, G., Marret, H., De Poncheville, L., et al. (2005) Ovarian and Fallopian Cancers. Processed of Gynecology. Medicine-Sciences, Flammarion Editions, Paris, 371-407.
[22] Sando, Z., Mboudou, E., Fouoguet, T.J., et al. (2010) Clinical and Pathological Profile of Ovarian Cancer in Yaounde (Cameroon). Wink in Mothers and Child Heath, 7, 1183.
[23] N’Dah, K.J., Effi, A.B., Yao, G.V., et al. (2010) Epidemiological and Histological Aspects of Burkitt’s Lymphoma in Ivory Coast. About 245 Cases Collected in 24 Years (1984-2007). African Journal of Pathology, 9, 17-25.
[24] Seigneurin, J.M., Fafi-Kremer, S., Baccard, M., et al. (2006) The Epstein-Barr Virus and Infection Markers. Training Manual. Medical Biology, 36, 48-50.
[25] Parkin, D.M., Whelan, S.L., Ferlay, J., et al. (1997) Cancer in Five Continents. Vol. VII, IARC Scientific Publications, Lyon, 143.
[26] Descamps, P., Catala, L., Lefebvre, C., et al. (2005) Endometrial Cancer. Processed of Gynecology, Medicine-Sciences, Flammarion Editions, Paris, 332-349.                                                                                         eww141110lx
[27] Belanger, H. and Roy, M. (1994) Vulvar Women’s Health. Edisem Editions FMOQ Malone, Saint-Hyacinthe, 711-716.