| Peer-Reviewed

The Impact of Middle Age on the Viability of Patients with Nonmalignant and Malignant Diseases

Received: 8 November 2014    Accepted: 28 November 2014    Published: 2 December 2014
Views:       Downloads:
Abstract

Deep myelosuppression, an officially sanctioned effect of non-selective cytotoxic cancer therapy, would be expected to be incompatible with mounting of a powerful host defense against spontaneous malignancy. To explore this theoretical difficulty, we used middle age as a natural model of a temporary decline in lymphocytopoiesis, caused by physiological thymus involution. The impact of middle age on the levels of death from nonmalignant and malignant diseases was analyzed retrospectively, using population health data from Europe (the European Network of Economic Policy Research Institutes, 1995); the UK (Statistics Team at the Cancer Research UK, and the Office for National Statistics cancer survival rates for 2007-2010), and the USA (National Center for Health Statistics, 1987-2007; National Vital Statistics System, 1999-2010; National Cancer Institute's Surveillance, Epidemiology, and End Results [SEER], 1992-2010). The rate of death and survival used to check whether the vectors of middle age-specific changes of these parameters are opposite or coincident in cancer patients and those with certain non-malignant somatic diseases. According the temporary trend on a middle- age portion of plot, the curves were graded negative or positive (+ = viability is not change or goes up; - = viability goes down).Comparisons of aggregate data showed that middle age exerted opposite effects on the health of those with cancer and non-malignant diseases. In middle age, serious health conditions, such as some cancers, are easier to treat, but the overall quality of life is reduced by various morbidities, especially infections. The comparing of the impact of middle age on the viability of patients with nonmalignant and malignant diseases in alternative terms of immunity or morphogenesis leads to recognition of trophic contribution of thymus into tumor development. By analogy, we assume that use of cytotoxic therapy can exert indirect benefit, thus compromising hemato- lymphocytopoiesis.

Published in Cancer Research Journal (Volume 2, Issue 6)
DOI 10.11648/j.crj.20140206.14
Page(s) 114-120
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Death Rates, Malignancy, Middle Age, Mielopoiesis, Populations

References
[1] A.L. Gruver, L.L. Hudson, and G.D. Sempowski, “Immunosenescence of ageing”, J Pathol, vol. 211(2), pp. 144–156, Jan 2007.
[2] H. E. Lynch, G. L. Goldberg, A. Chidgey, R. Boyd, and G. D. Sempowski, “Thymic involution and immune reconstitution,” Trends in Immunol, vol. 30(7), pp. 366–373, Jun 2009. doi:10.1016/j.it.2009.04.003
[3] U.S. Department of Health and Human Services, National Institute of Health, National Cancer Institute. “Blood/bone marrow,” in: Common terminology criteria for adverse events (CTCAE)/Version 3.0., AMGEN Oncology, p.4, August 2006. ctep.cancer.gov/.../electronic.../ctcaev3.pdf
[4] M. Akiyama and Y Kusunoki, “Immune function,“ in: Effects of A-bomb radiation on the human body, I..Shigematsu, C. Ito, N. Kamada, M. Akiyama, H. Sasaki, B. Harrison, Eds. Tokyo, Japan, Harwood academic publishers, Bunkodo Co., 1995, pp 290–306.
[5] J. G. Simpson, E. S. Gray, and J. S. Beck, “Age involution in the normal human adult thymus,” Clin Exp Immunol, vol. 19(2), pp. 261–265, Feb 1975. PMCID: PMC1538100
[6] F. Hakim, S. Memon, R. Cepeda, E. Jones, C. Chow, C. Kasten-Sportes, J. Odom, B. Vance, B. Christensen,. C. L. Mackall, and R. E Gress, “Age-dependent incidence, time course, and consequences of thymic renewal in adults,” J Clin Invest, vol. 115(4), pp. 930–939, March 2005. doi:10.1172/JCI22492. PMC 1064981.
[7] S. D. Gore, M. B. Kastan, and K. I. Civin, “Normal human bone marrow pre-courses that express terminal deoxynucleotidyl transferase include T-cell pre-courses and possible lymphoid stem cells,” Blood, vol. 77(8), pp. 1681-1690, April 1991. http://bloodjournal.hematologylibrary.org/content/77/8/1681
[8] M. A. Karamullin, A. E. Sosyukin, A. N. Shoutko, K. V. Nedoborski, and L. P. Ekimova, “Possible role of determined by age changes of the lymphopoiesis in the long-term morbidity levels of Chernobyl’ clean-up workers,” Meditsinskaya Radiologiya I Radiatsionnaya Bezopasnost, vol. 51(4), pp. 42–51, http://www.nuclearmedicine.ru/index.php/2010-06-07-07-49-12/2010-06-07-07-49-39/103-2010-07...
[9] M. Rifa'i, Y. Kawamoto, I. Nakashima, and H. Suzuki, (2004). “Essential roles of CD8+CD122+ regulatory T cells in the maintenance of T cell homeostasis,” J Exp Med, vol. 200 (9), pp. 1123-1134. doi: 10.1084/jem.20040395
[10] M. Fujiwara, T. Yonezawa, T. Arai, I. Yamamoto, and H. Ohtsuka, “Alterations with age in peripheral blood lymphocyte subpopulations and cytokine synthesis in beagles,” Veterinary Medicine: Research and Reports, vol. 3, pp. 79–84, August 2012. doi: http://dx.doi.org/10.2147/VMRR.S32590
[11] G Aubert and P. M. Lansdorp, “Telomeres and aging,” Physiol Rev, vol. 88, pp. 557-579, April 2008. doi: 10.1152/physrev.00026.2007
[12] J. M. Thuringer, and A. A. Katzberg, “The effect of age on mitosis in the human epidermis,” The Journal of Investigative Dermatology, vol. 3, pp. 35–39, November 1959. doi:10.1038/jid.1959.119
[13] A. N. Shutko, I. Akushevich, L. P. Ekimova, and M. A. Karamullin, “Variation of mortality risk among cancer patients and individualized therapy,” Vopr Onkol, vol. 56(4), pp. 430–434, Jul-August 2010. [In Russian]. PMID: 20968022
[14] K. Odunsi, and L. J. Old, “Tumor infiltrating lymphocytes: indicators of tumor related immune responses,” Canc Immun, vol. 7, pp.1–3, February 2007. PMCID: PMC 2935754
[15] National Vital Statistics System: Mortality Tables. LCWK1. 2010. http://www.cdc.gov/nchs/nvss/mortality_tables.htm.
[16] American Lung Association (ALA). Centres for Disease Control and Prevention, National Centre for Health Statistics, National Health Interview Survey. (2007). “Influenza and pneumonia – number of cases and rate per 1,000 population among youth and adults by race, sex and age, 2007,” in: Trends in pneumonia and influenza morbidity and mortality. ALA, research and program services epidemiology and statistics unit, p.16, (2008). http://drtenpenny.com/wp-content/uploads/2012/07/FLU-STATS vs PNEUMONIA 2008.pdf
[17] National Center for Health Statistics. Health, United States, 2010 “With special feature on death and dying,” Hyattsville, MD, 2011. www.cdc.gov/nchs/data/hus/hus10.pdf
[18] R. Layte, A. Nolan, B. Nolan, and T. Van Ourti (2005). “Health and morbidity by age and socio-economic characteristic,” European Network of Economic Policy Research Institutes (ENEPRI), ENEPRI research report 15, pp. 1-119, http://aei.pitt.edu/9488/2/9488.pdf;HTTP://WWW.ENEPRI.ORG
[19] The Statistics Team at Cancer Research UK “Cancer survival statistics: cancer research UK,” Accessed March 2014 in: www.cancerresearchuk.org/.../survival/england
[20] National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER 13, USA). [seer.cancer.gov/statistics/types/survival.html]
[21] Curve fitting project - pdf.io, pp.1-4.dynsys.uml.edu/tutorials/Regression.../curve_fit_proj_101905.pdf
[22] J. L. Loveland, (2011).Mathematical justification of introductory hypothesis tests and development of reference materials (M.Sc. (Mathematics)). Utah State University, Retrieved April 2013. http://digitalcommons.usu.edu/cgi/viewcontent.cgi?article=1014&context=gradreports
[23] J. Baba, S. Watanabe, Yu. Saida, T. Tanaka, T. Miyabayash, J. Koshio, K. Ichikawa, K. Nozaki, T. Koya, K. Deguchi, C. Tan, S. Miura, H. Tanaka, J. Tanaka, H. Kagamu, H. Yoshizawa, K. Nakata, and. I. Narita, “Depletion of radio-resistant regulatory T cells enhances antitumor immunity during recovery from lymphopenia,” Blood, vol. 120(12), pp. 2417-2427, July 2012. doi: 10.1182/blood-2012-02-411124 PMID:22806892
[24] E. L. Kachikwu, K. S. Iwamoto, Y. P. Liao, J. J. De Marco, N. Agazaryan, J. S. Economou, W. H. McBrige, and D. Schaue, “Radiation enhances regulatory T cell representation,” Int J Radiat Oncol Biol Phys, vol. 81(4), pp. 1128-1135, November 2011. doi: 10.1016/j.ijrobp.2010.09.034...
[25] L.-X. Wang, Y. Li, G. Yang, P.-V. Pang, D. Haley, E. B. Walker, W. J. Urba, and H.-M. Hu, (2010). “CD122+CD8+ Treg suppress vaccine-induced antitumor immune responses in lymphodepleted mice,” Eur J Immunol, vol. 40 (5), pp. 1375–1385, May 2010. doi: 10.1002/eji.200839210
[26] M. Kucia, J. Ratajczak, and M. Z. Ratajczak, “Bone marrow as a source of circulating CXCR4+ tissue- committed stem cells,” Biol Cell, vol. 97, pp. 133–146, February 2005. doi:10.1042/BC20040069
[27] Ch. Drapeau, Cracking the stem cell code: demystifying the most dramatic scientific breakthrough of our times. Hillsboro, Or, Goodwill Books USA, Sutton Hart Press / 1st. ed, 2010. http://www.amazon.com/Cracking-Stem-Cell-Code-Miraculous/dp/098102095X
[28] J. Hur, H.-M. Yang, C.-H. Yoon, C.-S. Lee, K.-W. Park, J.-H. Kim, T.-Y. Kim, J.-Y. Kim, H.-J. Kang, I.-H. Chae, B.-H. Oh, Y.-B. Park, and H.-S. Kim, “Identification of a novel role of T cells in postnatal vasculogenesis. Characterization of endothelial progenitor cell colonies,” Circulation, vol. 116(15), pp. 1671-1682, October 2007. PMID:17909106
[29] M. Roullet, S. M. F. Gheith, J. Mauger, J. M. Junkins-Hopkins, and J. K. Choi, “Percentage of γδ T cells in panniculitis by paraffin immunohistochemical analysis,” Am J Clin Pathol, vol. 131, pp. 820–826, 2009. doi: 10.1309/AJCPMG37MXKYPUBE
[30] A. N. Shoutko and L.P. Ekimova, “Abnormal tissue proliferation and life span variability in chronically irradiated dogs,” Radiat Environ Biophys, vol. 53(1), pp. 65-72, March 2014. doi: 10.1007/s00411-013-0504-7
[31] P. Starlinger, P. Brugger, C. Reiter, D. Schauer, S. Sommerfeldt, l D. Tamand, I. Kuehrer, S. F. Schoppmann, M. Gnant, and C. Brostjan, “Discrimination between circulating endothelial cells and blood cell populations with overlapping phenotype reveals distinct regulation and predictive potential in cancer therapy,” Neoplasia, vol. 13(10), pp. 980–990, September 2011. doi: 10.1593/neo.11916
[32] G. Beretta, Cancer chemotherapy regimens. Milano, Italy, Farmitalia Carlo Erba, 1983. opac.sbn.it/.../opaclib?..
[33] K.J. Propert and J.R. Anderson, “Assessing the effect of toxicity on prognosis: methods of analysis and interpretation,” JCO, vol. 6, pp. 868-870, month 1988. http://jco.ascopubs.org/content/6/5/868.full.pdf
[34] V. Malhotra and M. C. Perry, “Classical chemotherapy: mechanisms, toxicities and the therapeutic window,” Cancer Biol Ther, vol. 2(4 Suppl 1), pp. S2-4, Jul-Aug 2003. PMID:14508075
[35] W. L. Chen, Y. C. Luan, M. C. Shieh, S. T. Chen, H. T. Kung, K. L. Soong, Y. C. Yeh, T. S. Chou, S. H. Mong, J. T. Wu, C. P. Sun, W. P. Deng, M. F. Wu, and M. L. Shen, “Effects of Cobalt-60 exposure on health of Taiwan residents suggest new approach needed in radiation protection dose response,” Dose response, vol. 5(1), pp. 63–75, August 2007. doi: 10.2203/dose-response.06-105.Chen
[36] T. D. Luckey “The health effects of low-dose ionizing radiation,” Journal of American physicians and surgeons, vol. 13(2), pp.39-42, Summer 2008. www.jpands.org/vol13no2/luckey.pdf
Cite This Article
  • APA Style

    Alexei N. Shoutko, Lyudmila P. Ekimova. (2014). The Impact of Middle Age on the Viability of Patients with Nonmalignant and Malignant Diseases. Cancer Research Journal, 2(6), 114-120. https://doi.org/10.11648/j.crj.20140206.14

    Copy | Download

    ACS Style

    Alexei N. Shoutko; Lyudmila P. Ekimova. The Impact of Middle Age on the Viability of Patients with Nonmalignant and Malignant Diseases. Cancer Res. J. 2014, 2(6), 114-120. doi: 10.11648/j.crj.20140206.14

    Copy | Download

    AMA Style

    Alexei N. Shoutko, Lyudmila P. Ekimova. The Impact of Middle Age on the Viability of Patients with Nonmalignant and Malignant Diseases. Cancer Res J. 2014;2(6):114-120. doi: 10.11648/j.crj.20140206.14

    Copy | Download

  • @article{10.11648/j.crj.20140206.14,
      author = {Alexei N. Shoutko and Lyudmila P. Ekimova},
      title = {The Impact of Middle Age on the Viability of Patients with Nonmalignant and Malignant Diseases},
      journal = {Cancer Research Journal},
      volume = {2},
      number = {6},
      pages = {114-120},
      doi = {10.11648/j.crj.20140206.14},
      url = {https://doi.org/10.11648/j.crj.20140206.14},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.crj.20140206.14},
      abstract = {Deep myelosuppression, an officially sanctioned effect of non-selective cytotoxic cancer therapy, would be expected to be incompatible with mounting of a powerful host defense against spontaneous malignancy. To explore this theoretical difficulty, we used middle age as a natural model of a temporary decline in lymphocytopoiesis, caused by physiological thymus involution. The impact of middle age on the levels of death from nonmalignant and malignant diseases was analyzed retrospectively, using population health data from Europe (the European Network of Economic Policy Research Institutes, 1995); the UK (Statistics Team at the Cancer Research UK, and the Office for National Statistics cancer survival rates for 2007-2010), and the USA (National Center for Health Statistics, 1987-2007; National Vital Statistics System, 1999-2010; National Cancer Institute's Surveillance, Epidemiology, and End Results [SEER], 1992-2010). The rate of death and survival used to check whether the vectors of middle age-specific changes of these parameters are opposite or coincident in cancer patients and those with certain non-malignant somatic diseases. According the temporary trend on a middle- age portion of plot, the curves were graded negative or positive (+ = viability is not change or goes up; - = viability goes down).Comparisons of aggregate data showed that middle age exerted opposite effects on the health of those with cancer and non-malignant diseases. In middle age, serious health conditions, such as some cancers, are easier to treat, but the overall quality of life is reduced by various morbidities, especially infections. The comparing of the impact of middle age on the viability of patients with nonmalignant and malignant diseases in alternative terms of immunity or morphogenesis leads to recognition of trophic contribution of thymus into tumor development. By analogy, we assume that use of cytotoxic therapy can exert indirect benefit, thus compromising hemato- lymphocytopoiesis.},
     year = {2014}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - The Impact of Middle Age on the Viability of Patients with Nonmalignant and Malignant Diseases
    AU  - Alexei N. Shoutko
    AU  - Lyudmila P. Ekimova
    Y1  - 2014/12/02
    PY  - 2014
    N1  - https://doi.org/10.11648/j.crj.20140206.14
    DO  - 10.11648/j.crj.20140206.14
    T2  - Cancer Research Journal
    JF  - Cancer Research Journal
    JO  - Cancer Research Journal
    SP  - 114
    EP  - 120
    PB  - Science Publishing Group
    SN  - 2330-8214
    UR  - https://doi.org/10.11648/j.crj.20140206.14
    AB  - Deep myelosuppression, an officially sanctioned effect of non-selective cytotoxic cancer therapy, would be expected to be incompatible with mounting of a powerful host defense against spontaneous malignancy. To explore this theoretical difficulty, we used middle age as a natural model of a temporary decline in lymphocytopoiesis, caused by physiological thymus involution. The impact of middle age on the levels of death from nonmalignant and malignant diseases was analyzed retrospectively, using population health data from Europe (the European Network of Economic Policy Research Institutes, 1995); the UK (Statistics Team at the Cancer Research UK, and the Office for National Statistics cancer survival rates for 2007-2010), and the USA (National Center for Health Statistics, 1987-2007; National Vital Statistics System, 1999-2010; National Cancer Institute's Surveillance, Epidemiology, and End Results [SEER], 1992-2010). The rate of death and survival used to check whether the vectors of middle age-specific changes of these parameters are opposite or coincident in cancer patients and those with certain non-malignant somatic diseases. According the temporary trend on a middle- age portion of plot, the curves were graded negative or positive (+ = viability is not change or goes up; - = viability goes down).Comparisons of aggregate data showed that middle age exerted opposite effects on the health of those with cancer and non-malignant diseases. In middle age, serious health conditions, such as some cancers, are easier to treat, but the overall quality of life is reduced by various morbidities, especially infections. The comparing of the impact of middle age on the viability of patients with nonmalignant and malignant diseases in alternative terms of immunity or morphogenesis leads to recognition of trophic contribution of thymus into tumor development. By analogy, we assume that use of cytotoxic therapy can exert indirect benefit, thus compromising hemato- lymphocytopoiesis.
    VL  - 2
    IS  - 6
    ER  - 

    Copy | Download

Author Information
  • Medical Radiobiology Department, Federal Scientific Centre for Radiology and Surgical Technologies, Saint-Petersburg, Russian Federation

  • Medical Radiobiology Department, Federal Scientific Centre for Radiology and Surgical Technologies, Saint-Petersburg, Russian Federation

  • Sections