Therapy for Children’s Cardiac Dysfunction Associated with Cancer Treatment View PDF

*Ariel Pablo Lopez
Medicine, Universidad De Buenos Aires, Universidad De Buenos Aires, Universidad De Buenos Aires, Buenos Aries, Argentina

*Corresponding Author:
Ariel Pablo Lopez
Medicine, Universidad De Buenos Aires, Universidad De Buenos Aires, Universidad De Buenos Aires, Buenos Aries, Argentina
Email:aplopez@prensamedica.com.ar

Published on: 2022-04-18

Abstract

A major health burden for pediatric cancer patients is cardiovascular disease. The study of cardiovascular diseases that emerge because of cancer therapy, including their diagnosis, tracking, and management, is the emphasis of the emerging discipline of cardio-oncology [1]. Limited smoking, early cancer detection, and novel therapies, particularly molecularly targeted therapy and immunotherapy are just a few of the variables that improve cancer survival [2]. Cardiotoxicity is also increased by additional risk factors like concurrent radiotherapy or chemotherapy, lower age at onset, feminine gender, comorbidities, lifestyle factors, and hereditary factors like hemochromatosis gene mutations. Childhood cancer mortality rates have greatly increased in recent years, with 5-year overall survival rates now surpassing 80%. Anthracyclines prevent cancer cells from proliferating by blocking topoisomerases, DNA crosslinking, Ribosome replication, and DNA replication [3]. When given anthracyclines (doxorubicin and daunorubicin) at dosages greater than 300 mg/m2 , about 10% of infants experience side effects. Anthracycline exposure during childhood increases the risk of developing heart failure by 15 times and the risk of dying from cardiovascular illness by 8 times. Different pediatric cancer victims with cancer-related cardiovascular events appear clinically in different ways. congenital heart disease has become the most prevalent congenital malformation, ranges from mild heart failure to over heart failure, and its prevalence has stayed high in recent years [4], CTRCD (cancer treatment-related cardiac dysfunction) is a range of cardiac dysfunction spanning from overt indications and clinical symptoms of heart failure to asymptomatic heart injury identified only by elevated biomarkers like troponin or brain natriuretic peptide (BNP). An interdisciplinary strategy among doctors is necessary for secondary prevention after the end of cancer therapy, such as improved monitoring, suitable screening methods, and prophylactic treatment of cardiovascular complications. Prevention, risk management, and early diagnosis are crucial because cardiotoxicity progression is permanent. Several pediatric tumors are treated with anthracyclines, an anticancer medication. Damage to the heart is a significant anthracycline adverse effect that can cause either asymptomatic (non-complaint) or symptomatic (complaint) cardiac issues during and after cancer therapy. The Children’s Oncology Group’s Guidance for Long-Term Obey in Children, Adolescents, and Adolescents [5] offer comprehensive information for organizations interested in developing and enhancing long-term follow-up initiatives for children with cancer. It is significant to observe that there are variations in CV monitoring between centers, resulting in significant practice variance.

Keywords

Children’s Cardiac Dysfunction, Cancer Treatment, Cancer, Pediatric Cancer Patients

Methods

Cardiovascular Toxic Effects Pathophysiology in Cancer Patients

A higher risk of cardiotoxicity was seen in the Childhood Cancer Survival Study (CCSS) at smaller total doses of 250 mg/m2 as opposed to 400 mg/m2 . According to the CCSS, there may not be a secure number of anthracyclines for toddlers to take. In one trial, unlike that in grownups, there was no distinction between bolus and anthracyclines in children’s cardiotoxicity results [6]. Patients with pediatric cancer or those who have endured anthracycline-related heart issues should be able to make educated choices about their care based on high-quality proof of the advantages and disadvantages of treatment alternatives. We located two randomized studies comparing the effects of two various medications on two various patient populations. Enalapril, an ACE inhibitor, is one of these medications that improves heart function in pediatric cancer patients with asymptomatic anthracycline-induced cardiac issues in comparison to placebo in the short term. We located two randomized studies comparing the effects of two various medications on two various patient populations. Enalapril, an ACE inhibitor, is one of these medications that improves heart function in pediatric cancer patients with asymptomatic anthracycline-induced cardiac issues in comparison to placebo in the short term. Doxorubicin is the anthracycline that is most frequently used to treat childhood cancer. It inhibits cellular proliferation and function, alters deoxyribonucleic acid regulation in cardiac tissue, and acts as an anticancer agent by interacting to topoisomerase II. By chemically reacting with Fe+2, doxorubicin causes lipid peroxidation in cells and mitochondrial membranes. It also causes mortality, mitochondria damage to DNA damage, and ecological collapse by releasing reactive oxygen species (ROS) [7]. There is no treatment for cancer that is completely safe, and all chemotherapy drugs can result in long-term cardiotoxicity [8]. Cardiotoxicity can be brought on by alkylating substances like cisplatin and carboplatin that enhance the generation of ROS and interfere with the antioxidant system [9]. Long-term cardiology follow-up is crucial because cisplatin and carboplatin can also cause late-onset CTRCD [10]. Cardiomyocyte apoptosis, inflammation, endothelial dysfunction, calcium dysregulation, and mitochondrial damage have all been linked to cyclophosphamide and have been shown to cause high blood pressure, cardiomyopathy, myocardial infarction, arrhythmias, and eventually heart failure. Several pediatric cancer patients are being treated with targeted cancer therapy. The processes driving cancer development have improved immune checkpoint inhibitors, also known as ICIs, such as ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, and durvalumab, as well as tyrosine kinase inhibitors, or TKIs, are used as treatment (imatinib, dasatinib, bevacizumab, bosutinib, sunitinib, pro sornanitinib, pro sornanitinib). Hypoxia-inducible factor 1-alpha (HIF1) is a protein that is targeted by vascular endothelial growth factor inhibitors, which results in myocardial hypoxia and cardiac failure. Refractory blood malignancies and acute lymphoblastic leukemia are being treated with cell treatments such chimeric antigen receptor T (CAR-T) cells. The carcinogenesis caused by CAR-T cells results from the direct cell-mediated lysis of cancer cells as well as a rise in serum potassium and uric acid. Syndrome of lysis six months after receiving CAR-T cell treatment, Burstein et al. found that 25% of children had hypotension and 10% had diminished cardiac function [11].

Heart Failure and Myocarditis in Oncology

Systolic or diastolic heart dysfunction is a common pathological defect in all CTRCDs. Cyclophosphamide, 5-FU, cisplatin, and RT are examples of secondary modifications that can cause inflammatory myocarditis or microvascular damage and fibrosis that causes cardiac dysfunction (immune checkpoint inhibitors and RT). Left ventricular (LV) dysfunction and heart failure are categorized into five categories by the National Cancer Institute as standard terminological criteria for cancer-related adverse outcomes in adults. Grade 1 biomarker increase, or imaging abnormality has no symptoms. Mild and moderate exertion-related symptoms were categorized into Grade 2 and Grade 3, respectively. Grade 4 symptoms are severe and life-threatening and require hemodynamic care, whereas Grade 5 symptoms are fatal. 62% of childhood cancer survivors have a major or life-threatening condition such a stroke, heart failure, or renal failure, and 27% of them have at least one chronic illness [12]. Anthracyclines’ ability to produce cardiotoxicity in children has been thoroughly researched. Nevertheless, at dosages as high as 60 mg/m2 , cardiovascular illness in children has been shown [13]. Chemotherapy causes a number of ongoing changes, including a rise in cardiac biomarkers, regional myocardial deformation (abnormal tension), and asymptomatic LV systolic or diastolic dysfunction, which ultimately result in heart failure [14]. Based on the emergence of clinical signs of persistent cardiotoxicity, childhood cancer survivors can be categorized into two categories. The second subtype develops later, more than a year after chemotherapy, and is often progressive. The first subtype manifests early, within a year following treatment. With a mean follow-up of 11.8 years, children receiving doxorubicin have lower LV wall thickness in proportion to somatic development when compared to untreated children [15]. Since injured myocytes fail to recover and are replaced by interstitial fibrosis following doxorubicin therapy, cardiac dysfunction persists and worsens [16]. Although LV mass and cavity size steadily decline in relation to body size, progressive restrictive cardiomyopathy is often permanent and is a long-term risk factor for heart failure with maintained EF in pediatric cancer survivors [17].

Diagnostic and Risk Analysis

Careful assessment of cardiovascular risk factors previously, all through, and then after cancer treatment with basic history, diagnostic and laboratory tests for cardiac biomarkers, and screening tests is required for the detection of high-risk patients and diagnosis  of cardiotoxicity in children before symptoms of heart failure appear (Figure 1). Cardiotoxicity risk is influenced by the patient’s cancer diagnosis, the type of therapy they got, and risky health habits including smoking, drinking, using drugs (such as cocaine, diet pills, and ephedra), eating poorly, being sedentary, and having comorbid conditions. existing cardiomyopathy, congenital cardiac disease, high blood pressure, hyperlipidemia, diabetes, and obesity [18]. Notably the Childhood Cancer Survivor Study have revealed characteristics that raise the risk of cardiotoxicity, such as younger patient age (5 years), smoking, and obesity. Trisomy 21, total anthracycline dosage, female gender, African American race, concomitant radiation, heart illness, premodern radiation procedures (before 1975), and length of time since therapy [19]. Also, there is proof that cancer patients already have subclinical cardiac impairment prior to receiving cancer therapy, which highlights the need of evaluating patients’ heart health prior to receiving cancer treatment [20]. In patients undergoing anticancer therapy who do not have CAD, elevated levels of cardiac troponin T, BNP, or NT-pro-BNP suggest subclinical cardiac injury and are linked to decreased LV function [21, 22]. Lower LV mass, wall thickness, and echocardiographic remodel five years after anthracycline treatment is related with cardiac damage (measured by blood cardiac troponin T 99th percentile) [23]. Serial biomarker screening is crucial for CTRCD adopt and prevent complications in childhood cancer survivors. A biomarker identified as galectin-3 has been linked to cardiac fibrosis and inflammation, whereas ST2, also known as soluble interleukin-1 receptor-like elevated myeloperoxidase was demonstrated to predict cardiotoxicity [24]. Exosomes have recently been discovered to be useful indicators of doxycycline-induced cardiotoxicity [25]. Exosomes are a subpopulation of extracellular vesicles that influence a number of pathophysiological processes.

Conclusion

Decreased exercise capacity is the greatest indicator of a modest deterioration in cardiac health for long-term cancer victims receiving cardiotoxic medication. Surgical intervention improves the quality of life and health in children with coronary artery disease, in addition to treating congenital abnormalities. To enhance the quality of life connected to health, children with coronary heart disease should be identified and treated as soon as feasible. Cardiotoxicity related to cancer treatment has the potential to be systemic, chronic, and progressive, and it may go unnoticed by doctors. One significant hypothesis is that chronic cardiotoxicity may be linked to persistent mitochondrial damage. As the number of cancer survivors rises, so does the health burden brought on by cardiovascular disease. Continuous follow-up is vital as the community of survivors of childhood cancer matures, and being able to provide quality care will help us do better. The healthcare system in the US is evolving, somewhat unsteady, and politically driven. It is a system that is experiencing a crisis, and just a little bit of funding has been set aside to avoid it. Lifelong follow-up is necessary for childhood cancer survivors, especially for those who are at high risk for developing CTRCD.

References

  1. Kostakou PM, Kouris NT, Kostopoulos VS, Damaskos DS, Olympios CD (2019) Cardio-oncology: a new and developing sector of research and therapy in the field of cardiology. Heart Fail Rev 24: 91-100. https://doi.org/10.1007/s10741-018-9731-y
  2. Siegel RL, Miller KD, Fuchs HE, Jemal A (2022) Cancer statistics, 2022. CA Cancer J Clin 72: 7-33. https://doi.org/10.3322/caac.21708
  3. Ewer MS, Ewer SM (2015) Cardiotoxicity of anticancer treatments. Nat Rev Cardiol 12: 547-558. https://doi.org/10.1038/nrcardio.2015.65
  4. Hoffman JI (2013) The global burden of congenital heart disease. Cardiovasc J Africa 24: 141-145. https://doi.org/10.5830/CVJA-2013-028
  5. Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent, and Young Adult Cancers.
  6. Lipshultz SE, Miller TL, Lipsitz SR, Neuberg DS, Dahlberg SE, et al. (2012) Continuous versus bolus infusion of doxorubicin in children with ALL: long-term cardiac outcomes. Pediatrics 130: 1003-1011. https://doi.org/10.1542/peds.2012-0727
  7. Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, et al. (2012) Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med 18: 1639-1642. https:// doi.org/10.1038/nm.2919
  8. Lipshultz SE, Rifai N, Dalton VM, Levy DE, Silverman LB, et al. (2004) The effect of dexrazoxane on myocardial injury in doxorubicin-treated children with acute lymphoblastic leukemia. N Eng J Med 351: 145-153. https://doi.org/10.1056/ NEJMoa035153
  9. Akam-Venkata J, Franco VI, Lipshultz SE (2016) Late cardiotoxicity: issues for childhood cancer survivors. Curr Treat Options Cardiovasc Med 18: 1-23. https://doi. org/10.1007/s11936-016-0466-6
  10. Zhang X, Zhu Y, Dong S, Zhang A, Lu Y, et al. (2019) Role of oxidative stress in cardiotoxicity of antineoplastic drugs. Life Sci 232: 116526. https://doi.org/10.1016/j. lfs.2019.06.001
  11. Burstein DS, Maude S, Grupp S, Griffis H, Rossano J, et al. (2018) Cardiac profile of chimeric antigen receptor T cell therapy in children: a single-institution experience. Biol Blood Marrow Transplant 24: 1590-1595. https://doi.org/10.1016/j.bbmt.2018.05.014
  12. Oeffinger KC, Mertens AC, Sklar CA, Kawashima T, Hudson MM, et al. (2006) Chronic health conditions in adult survivors of childhood cancer. N Eng J Med 355: 1572-1582. https://doi.org/10.1056/NEJMsa060185
  13. Ganame J, Claus P, Eyskens B, Uyttebroeck A, Renard M, et al. (2007) Acute cardiac functional and morphological changes after Anthracycline infusions in children. Am J Cardiol 99: 974-977. https://doi.org/10.1016/j.amjcard.2006.10.063
  14. Nicol M, Baudet M, Cohen-Solal A (2019) Subclinical left ventricular dysfunction during chemotherapy. Card Fail Rev 5: 31. https://doi.org/10.15420/cfr.2018.25.1
  15. Lipshultz SE, Lipsitz SR, Sallan SE, Dalton VM, Mone SM, et al. (2005) Chronic progressive cardiac dysfunction years after doxorubicin therapy for childhood acute lymphoblastic leukemia. J Clin Oncol 23: 2629-2636. https://doi.org/10.1200/ JCO.2005.12.121
  16. Lipshultz SE, Colan SD, Gelber RD, Perez-Atayde AR, Sallan SE, et al. (1991) Late cardiac effects of doxorubicin therapy for acute lymphoblastic leukemia in childhood. N Eng J Med 324: 808-815. https://doi.org/10.1056/NEJM199103213241205
  17. Das B, Deshpande S, Akam-Venkata J, Shakti D, Moskowitz W, et al. (2022) Heart failure with preserved ejection fraction in children. Pediatr Cardiol 44: 513-529. https:// doi.org/10.1007/s00246-022-02960-7
  18. Mulrooney DA, Armstrong GT, Huang S, Ness KK, Ehrhardt MJ, et al. (2016) Cardiac outcomes in adult survivors of childhood cancer exposed to cardiotoxic therapy: a cross-sectional study. Ann Intern Med 164: 93-101. https://doi.org/10.7326/M15-0424
  19. Lee JW, Aminkeng F, Bhavsar AP, Shaw K, Carleton BC, et al. (2014) The emerging era of pharmacogenomics: current successes, future potential, and challenges. Clin Genet 86: 21-28. https://doi.org/10.1111/cge.12392
  20. Yingchoncharoen T, Agarwal S, Popovi? ZB, Marwick TH (2013) Normal ranges of left ventricular strain: a meta-analysis. J Am Soc Echocardiogr 26: 185-191. https://doi. org/10.1016/j.echo.2012.10.008
  21. Cardinale D, Sandri MT, Colombo A, Colombo N, Boeri M, et al. (2004) Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing highdose chemotherapy. Circulation 109: 2749-2754. https://doi.org/10.1161/01. CIR.0000130926.51766.CC
  22. Lipshultz SE, Landy DC, Lopez-Mitnik G, Lipsitz SR, Hinkle AS, et al. (2012) Cardiovascular status of childhood cancer survivors exposed and unexposed to cardiotoxic therapy. J Clin Oncol 30: 1050-1057. https://doi.org/10.1200/ JCO.2010.33.7907
  23. Kellman P, Wilson JR, Xue H, Ugander M, Arai AE (2012) Extracellular volume fraction mapping in the myocardium, part 1: evaluation of an automated method. J Cardiovasc Magn Reson 14: 1-11. https://doi.org/10.1186/1532-429X-14-63
  24. Kremer LCM, Van Dalen EC, Offringa M, Ottenkamp J, Voute PA (2001) Anthracyclineinduced clinical heart failure in a cohort of 607 children: long-term follow-up study. J Clin Oncol 19: 191-196. https://doi.org/10.1200/JCO.2001.19.1.191
  25. Oikawa M, Yaegashi D, Yokokawa T, Misaka T, Sato T, et al. (2022) D-Dimer is a predictive factor of cancer therapeutics-related cardiac dysfunction in patients treated with cardiotoxic chemotherapy. Front Cardiovasc Med 8: 1-7. https://doi.org/10.3389/ fcvm.2021.807754
  26. Hegazy M, Ghaleb S, Das BB (2023) Diagnosis and management of cancer treatmentrelated cardiac dysfunction and heart failure in children. Children 10: 149. https://doi. org/10.3390/children1001014
scroll up