Cardiovascular and oncological diseases — focus on modifiable risk factors and modern pathogenetic aspects

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

Currently, in modern medicine, there has been a sharp increase in interest in the combined pathology of cardiovascular diseases (CVD) with other nosologies, such as diabetes mellitus, chronic kidney disease, and oncopathology. Over the past decades, there have been large positive shifts in survival in this cohort of patients. An integral specialty of cardio-oncology is rapidly developing, which is aimed at improving the results of treatment of oncological patients. Due to increased life expectancy and improved long-term prognosis in cancer patients, in particular, the adverse effects of anticancer therapy have attracted great attention from researchers. The causal relationship between cancer and CVD can be partly explained by a common profile of modifiable and non-modifiable risk factors. There is also strong evidence that a systemic inflammatory response is a common pathophysiological determinant of cancer and CVD. General risk factors for development and progression, as well as pathogenetic mechanisms, contribute to high mortality rates. Increasing knowledge of pathophysiological mechanisms will help elucidate the general molecular aspects of carcinogenesis and cardiovascular disease. This review presents general modifiable risk factors and current pathogenetic aspects of cardiovascular and oncological diseases.

Full Text

Restricted Access

About the authors

Jurij I. Buziashvili

A.N. Bakulev National Medical Research Center of Cardiovascular Surgery

Email: buziashvili@yandex.ru
ORCID iD: 0000-0001-7016-7541
SPIN-code: 2856-3356

MD, PhD, Professor, Academician of the RAS

Russian Federation, Moscow

Ivan S. Stilidi

N.N. Blokhin National Medical Research Center of Oncology

Email: director@ronc.ru
ORCID iD: 0000-0002-5229-8203
SPIN-code: 9622-7106

MD, PhD, Professor, Academician of the RAS

Russian Federation, Moscow

Simon T. Mackeplishvili

A.N. Bakulev National Medical Research Center of Cardiovascular Surgery

Email: simonmats@yahoo.com
ORCID iD: 0000-0002-5670-167X
SPIN-code: 2827-1317

MD, PhD, Professor, associate member RAS

Russian Federation, Moscow

El’mira U. Asymbekova

A.N. Bakulev National Medical Research Center of Cardiovascular Surgery

Email: elmasym@gmail.com
ORCID iD: 0000-0002-5422-2069
SPIN-code: 8108-3978

MD, PhD

Russian Federation, Moscow

Elvina F. Tugeeva

A.N. Bakulev National Medical Research Center of Cardiovascular Surgery

Email: Elvina.tugeeva@yandex.ru
ORCID iD: 0000-0003-1751-4924
SPIN-code: 3662-8775

MD, PhD

Russian Federation, Moscow

Elena V. Artamonova

N.N. Blokhin National Medical Research Center of Oncology; Pirogov Russian National Research Medical University; Moscow Regional Research and Clinical Institute (MONIKI)

Email: artamonovae@mail.ru
ORCID iD: 0000-0001-7728-9533
SPIN-code: 2483-6309

MD, PhD

Russian Federation, Moscow; Moscow; Moscow

Firdavsdzhon R. Akildzhonov

A.N. Bakulev National Medical Research Center of Cardiovascular Surgery

Author for correspondence.
Email: firdavs96_tths@mail.ru
ORCID iD: 0000-0002-1675-4216
Russian Federation, Moscow

Alier A. Rakhmanzhanov

A.N. Bakulev National Medical Research Center of Cardiovascular Surgery

Email: alier.raxmanzhanov@mail.ru
ORCID iD: 0000-0002-7800-9078
Russian Federation, Moscow

Elena Z. Golukhova

A.N. Bakulev National Medical Research Center of Cardiovascular Surgery

Email: egoluhova@bakulev.ru
ORCID iD: 0000-0002-6252-0322
SPIN-code: 9334-5672

MD, PhD, Professor, Academician of the RAS

Russian Federation, Moscow

References

  1. Sturgeon K, Deng L, Bluethmann S. A population-based study of cardiovascular disease mortality risk in US cancer patients. Eur Heart J. 2019;40(48):3889–3897. doi: https://doi.org/10.1093/eurheartj/ehz766
  2. Ngo D, Williams T, Horder S. Factors Associated with Adverse Cardiovascular Events in Cancer Patients Treated with Bevacizumab. J Clin Med. 2020;9(8):2664. doi: https://doi.org/10.3390/jcm9082664
  3. Al-Kindi S, Oliveira G. Prevalence of Preexisting Cardiovascular Disease in Patients with Different Types of Cancer: The Unmet Need for Onco-Cardiology. Mayo Clin Proc. 2016;91(1):81–83. doi: https://doi.org/10.1016/j.mayocp.2015.09.009
  4. Han X, Zhou Y, Liu W. Precision cardio-oncology: understanding the cardiotoxicity of cancer therapy. NPJ Precis Oncol. 2017;1(1):31. doi: https://doi.org/10.1038/s41698-017-0034-x
  5. Sung H, Ferlay J, Siegel R. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–249. doi: https://doi.org/10.3322/caac.21660
  6. Thun M, DeLancey J, Center M, et al. The global burden of cancer: priorities for prevention. Carcinogenesis. 2010;31(1):100–110. doi: https://doi.org/10.1093/carcin/bgp263
  7. Nguyen P, Saito E, Katanoda K. Long-Term Projections of Cancer Incidence and Mortality in Japan and Decomposition Analysis of Changes in Cancer Burden, 2020–2054: An Empirical Validation Approach. Cancers (Basel). 2022;14(24):6076. doi: https://doi.org/10.3390/cancers14246076
  8. Васюк Ю.А., Гендлин Г.Е., Емелина Е.И. Согласованное мнение российских экспертов по профилактике, диагностике и лечению сердечно-сосудистой токсичности противоопухолевой терапии // Российский кардиологический журнал. — 2021. — Т. 26. — № 9. — 4703. doi: https://doi.org/10.15829/1560-4071-2021-4703 [Vasyuk Yu, Gendlin G, Emelina EI. Consensus statement of Russian experts on the prevention, diagnosis and treatment of cardiotoxicity of anticancer therapy. Russian Journal of Cardiology. 2021;26(9):4703. (In Russ.)] doi: https://doi.org/10.15829/1560-4071-2021-4703
  9. Lyon A, López-Fernández T, Couch L. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J. 2022;43(41):4229–4361. doi: https://doi.org/10.1093/eurheartj/ehac244
  10. Khanna A, Pequeno P, Gupta S. Increased Risk of All Cardiovascular Disease Subtypes Among Childhood Cancer Survivors: Population-Based Matched Cohort Study. Circulation. 2019;140(12):1041–1043. doi: https://doi.org/10.1161/CIRCULATIONAHA.119.041403
  11. Clèries R, Ameijide A, Buxó M. Ten-Year Probabilities of Death Due to Cancer and Cardiovascular Disease among Breast Cancer Patients Diagnosed in North-Eastern Spain. Int J Environ Res Public Health. 2022;20(1):405. doi: https://doi.org/10.3390/ijerph20010405
  12. Zamorano J, Gottfridsson C, Asteggiano R. The cancer patient and cardiology. Eur J Heart Fail. 2020;22(12):2290–2309. doi: https://doi.org/10.1002/ejhf.1985
  13. Quoc Lam B, Shrivastava S, Shrivastava A, et al. The Impact of obesity and diabetes mellitus on pancreatic cancer: Molecular mechanisms and clinical perspectives. J Cell Mol Med. 2020;24(14):7706–7716. doi: https://doi.org/10.1111/jcmm.15413
  14. Krupa-Kotara K, Dakowska D. Impact of obesity on risk of cancer. Cent Eur J Public Health. 2021;29(1):38–44. doi: https://doi.org/10.21101/cejph.a5913
  15. Du X, Hidayat K, Shi B. Abdominal obesity and gastroesophageal cancer risk: systematic review and meta-analysis of prospective studies. Biosci Rep. 2017;37(3):BSR20160474. doi: https://doi.org/10.1042/BSR20160474
  16. Shang L, Hattori M, Fleming G. Impact of post-diagnosis weight change on survival outcomes in Black and White breast cancer patients. Breast Cancer Res. 2021;23(1):18. doi: https://doi.org/10.1186/s13058-021-01397-9
  17. Tangvarasittichai S, Pongthaisong S, Tangvarasittichai O. Tumor Necrosis Factor-Α, Interleukin-6, C-Reactive Protein Levels and Insulin Resistance Associated with Type 2 Diabetes in Abdominal Obesity Women. Indian J Clin Biochem. 2016;31(1):68–74. doi: https://doi.org/10.1007/s12291-015-0514-0
  18. Poetsch M, Strano A, Guan K. Role of Leptin in Cardiovascular Diseases. Front Endocrinol (Lausanne). 2020;11:354. doi: https://doi.org/10.3389/fendo.2020.00354
  19. Ellulu M, Patimah I, Khaza’ai H, et al. Obesity and inflammation: the linking mechanism and the complications. Arch Med Sci. 2017;13(4):851–863. doi: https://doi.org/10.5114/aoms.2016.58928
  20. Xu J, Lin H, Wu G, et al. IL-6/STAT3 Is a Promising Therapeutic Target for Hepatocellular Carcinoma. Front Oncol. 2021;11:760971. doi: https://doi.org/10.3389/fonc.2021.760971
  21. Eketunde A. Diabetes as a Risk Factor for Breast Cancer. Cureus. 2020;12(5):e8010. doi: https://doi.org/10.7759/cureus.8010
  22. Xu C, Zhu H, Zhu Y. Diabetes and cancer: Associations, mechanisms, and implications for medical practice. World J Diabetes. 2014;5(3):372–380. doi: https://doi.org/10.4239/wjd.v5.i3.372
  23. Hormati A, Hajrezaei Z, Jazi K, et al. Gastrointestinal and Pancratohepatobiliary Cancers: A Comprehensive Review on Epidemiology and Risk Factors Worldwide. Middle East J Dig Dis. 2022;14(1):5–23. doi: https://doi.org/10.34172/mejdd.2022.251
  24. Gu L, Ma G, Li C, et al. New insights into the prognosis of intraocular malignancy: Interventions for association mechanisms between cancer and diabetes. Front Oncol. 2022;12:958170. doi: https://doi.org/10.3389/fonc.2022.958170
  25. Travis R, Appleby P., Martin R. A Meta-analysis of Individual Participant Data Reveals an Association between Circulating Levels of IGF-I and Prostate Cancer Risk. Cancer Res. 2016;76(8):2288–2300. doi: https://doi.org/10.1158/0008-5472.CAN-15-1551
  26. Nounu A, Kar S, Relton C, Richmond R. Sex steroid hormones and risk of breast cancer: a two-sample Mendelian randomization study. Breast Cancer Res. 2022;24(1):66. doi: https://doi.org/10.1186/s13058-022-01553-9
  27. Ba Z, Xiao Y, He M. Risk Factors for the Comorbidity of Hypertension and Renal Cell Carcinoma in the Cardio-Oncologic Era and Treatment for Tumor-Induced Hypertension. Front Cardiovasc Med. 2022;9:810262. doi: https://doi.org/10.3389/fcvm.2022.810262
  28. El Hadri K, Smith R, Duplus E, El Amri C. Inflammation, Oxidative Stress, Senescence in Atherosclerosis: Thioredoxine-1 as an Emerging Therapeutic Target. Int J Mol Sci. 2021;23(1):77. doi: https://doi.org/10.3390/ijms23010077
  29. Angel-Korman A, Rapoport V, Leiba A. The Relationship between Hypertension and Cancer. Isr Med Assoc J. 2022;24(3):165–169.
  30. Grossman E, Messerli F, Boyko V, Goldbourt U. Is there an association between hypertension and cancer mortality? Am J Med. 2002;112(6):479–486. doi: https://doi.org/10.1016/s0002-9343(02)01049-5
  31. Skorupan N, Palestino Dominguez M, Ricci S, Alewine C. Clinical Strategies Targeting the Tumor Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel). 2022;14(17):4209. doi: https://doi.org/10.3390/cancers14174209
  32. Cedó L, Reddy S, Mato E, et al. HDL and LDL: Potential New Players in Breast Cancer Development. J Clin Med. 2019;8(6):853. doi: https://doi.org/10.3390/jcm8060853
  33. Larsson S, Carter P, Kar S. Smoking, alcohol consumption, and cancer: A mendelian randomisation study in UK Biobank and international genetic consortia participants. PLoS Med. 2020;17(7):e1003178. doi: https://doi.org/10.1371/journal.pmed.1003178
  34. Greten F, Grivennikov S. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity. 2019;51(1):27–41. doi: https://doi.org/10.1016/j.immuni.2019.06.025
  35. Perillo B, Di Donato M, Pezone A. ROS in cancer therapy: the bright side of the moon. Exp Mol Med. 2020;52(2):192–203. doi: https://doi.org/10.1038/s12276-020-0384-2
  36. Ridker P. From C-Reactive Protein to Interleukin-6 to Interleukin-1: Moving Upstream To Identify Novel Targets for Atheroprotection. Circ Res. 2016;118(1):145–156. doi: https://doi.org/10.1161/CIRCRESAHA.115.306656
  37. Johnson D, O’Keefe R, Grandis J. Targeting the IL-6/JAK/STAT3 signaling axis in cancer. Nat Rev Clin Oncol. 2018;15(4):234–248. doi: https://doi.org/10.1038/nrclinonc.2018.8
  38. Ya G, Ren W, Qin R, et al. Role of myeloid-derived suppressor cells in the formation of pre-metastatic niche. Front Oncol. 2022;12:975261. doi: https://doi.org/10.3389/fonc.2022.975261
  39. Crossman D, Rothman A. Interleukin-1 beta inhibition with canakinumab and reducing lung cancer-subset analysis of the canakinumab anti-inflammatory thrombosis outcome study trial (CANTOS). J Thorac Dis. 2018;10(Suppl 26):S3084–S3087. doi: https://doi.org/10.21037/jtd.2018.07.50
  40. Lust J, Lacy M, Zeldenrust S. Induction of a chronic disease state in patients with smoldering or indolent multiple myeloma by targeting interleukin 1{beta}-induced interleukin 6 production and the myeloma proliferative component. Mayo Clin Proc. 2009;84(2):114–122. doi: https://doi.org/10.4065/84.2.114
  41. Nguyen M, Bui K, Scholta T. Targeting interleukin 6 signaling by monoclonal antibody siltuximab on cholangiocarcinoma. J Gastroenterol Hepatol. 2021;36(5):1334–1345. doi: https://doi.org/10.1111/jgh.15307
  42. Kuo M, Chang S, Hsieh M. Colchicine Significantly Reduces Incident Cancer in Gout Male Patients: A 12-Year Cohort Study. Medicine (Baltimore). 2015;94(50):e1570. doi: https://doi.org/10.1097/MD.0000000000001570
  43. Carreira R, Lee P, Gottlieb R. Mitochondrial therapeutics for cardioprotection. Curr Pharm Des. 2011;17(20):2017–2035. doi: https://doi.org/10.2174/138161211796904777
  44. Roth K, Mambetsariev I, Kulkarni P, Salgia R. The Mitochondrion as an Emerging Therapeutic Target in Cancer. Trends Mol Med. 2020;26(1):119–134. doi: https://doi.org/10.1016/j.molmed.2019.06.009
  45. Bowman R, Busque L, Levine R. Clonal Hematopoiesis and Evolution to Hematopoietic Malignancies. Cell Stem Cell. 2018;22(2):157–170. doi: https://doi.org/10.1016/j.stem.2018.01.011
  46. Poller W, Nahrendorf M, Swirski F. Hematopoiesis and Cardiovascular Disease. Circ Res. 2020;126(8):1061–1085. doi: https://doi.org/10.1161/CIRCRESAHA.120.315895
  47. Libby P, Sidlow R, Lin A. Clonal Hematopoiesis: Crossroads of Aging, Cardiovascular Disease, and Cancer: JACC Review Topic of the Week. J Am Coll Cardiol. 2019;74(4):567–577. doi: https://doi.org/10.1016/j.jacc.2019.06.007
  48. Jaiswal S, Natarajan P, Silver A. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med. 2017;377(2):111–121. doi: https://doi.org/10.1056/NEJMoa1701719
  49. Senguttuvan N, Subramanian V, Venkatesan V, et al. Clonal hematopoiesis of indeterminate potential (CHIP) and cardiovascular diseases-an updated systematic review. J Genet Eng Biotechnol. 2021;19(1):105. doi: https://doi.org/10.1186/s43141-021-00205-3
  50. Pascual-Figal D, Bayes-Genis A, Díez-Díez M. Clonal Hematopoiesis and Risk of Progression of Heart Failure With Reduced Left Ventricular Ejection Fraction. J Am Coll Cardiol. 2021;77(14):1747–1759. doi: https://doi.org/10.1016/j.jacc.2021.02.028

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Figure 1. Types of inflammation in cancer: timing and inducers (adapted from [34])

Download (220KB)

Copyright (c) 2023 "Paediatrician" Publishers LLC



This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies