51
|
Lee DE, Brown JL, Rosa‐Caldwell ME, Perry RA, Brown LA, Haynie WS, Washington TA, Wiggs MP, Rajaram N, Greene NP. Cancer-induced Cardiac Atrophy Adversely Affects Myocardial Redox State and Mitochondrial Oxidative Characteristics. JCSM RAPID COMMUNICATIONS 2021; 4:3-15. [PMID: 33693448 PMCID: PMC7939061 DOI: 10.1002/rco2.18] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
UNLABELLED Cachexia presents in 80% of advanced cancer patients; however, cardiac atrophy in cachectic patients receives little attention. This cardiomyopathy contributes to increased occurrence of adverse cardiac events compared to age-matched population norms. Research on cardiac atrophy has focused on remodeling; however, alterations in metabolic properties may be a primary contributor. PURPOSE Determine how cancer-induced cardiac atrophy alters mitochondrial turnover, mitochondrial mRNA translation machinery and in-vitro oxidative characteristics. METHODS Lewis lung carcinoma (LLC) tumors were implanted in C57BL6/J mice and grown for 28days to induce cardiac atrophy. Endogenous metabolic species, and markers of mitochondrial function were assessed. H9c2 cardiomyocytes were cultured in LLC-conditioned media with(out) the antioxidant MitoTempo. Cells were analyzed for ROS, oxidative capacity, and hypoxic resistance. RESULTS LLC heart weights were ~10% lower than controls. LLC hearts demonstrated ~15% lower optical redox ratio (FAD/FAD+NADH) compared to PBS controls. When compared to PBS, LLC hearts showed ~50% greater COX-IV and VDAC, attributed to ~50% lower mitophagy markers. mt-mRNA translation machinery was elevated similarly to markers of mitochondrial content. mitochondrial DNA-encoded Cytb was ~30% lower in LLC hearts. ROS scavengers GPx-3 and GPx-7 were ~50% lower in LLC hearts. Treatment of cardiomyocytes with LLC-conditioned media resulted in higher ROS (25%), lower oxygen consumption rates (10% at basal, 75% at maximal), and greater susceptibility to hypoxia (~25%) -- which was reversed by MitoTempo. CONCLUSION These results substantiate metabolic cardiotoxic effects attributable to tumor-associated factors and provide insight into interactions between mitochondrial mRNA translation, ROS mitigation, oxidative capacity and hypoxia resistance.
Collapse
Affiliation(s)
- David E. Lee
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
- Laboratory for Functional Optical Imaging and Spectroscopy, Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Jacob L. Brown
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
| | - Megan E. Rosa‐Caldwell
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
| | - Richard A. Perry
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
| | - Lemuel A. Brown
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
| | - Wesley S. Haynie
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
| | - Tyrone A. Washington
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
| | - Michael P. Wiggs
- Department of Health and Kinesiology, University of Texas at Tyler, Tyler, Texas, USA
| | - Narasimhan Rajaram
- Laboratory for Functional Optical Imaging and Spectroscopy, Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Nicholas P. Greene
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
52
|
Miyagawa Y, Nukaga S, Mori T, Fujiwara-Tani R, Fujii K, Mori S, Goto K, Kishi S, Sasaki T, Nakashima C, Ohmori H, Kawahara I, Luo Y, Kuniyasu H. Evaluation of cancer-derived myocardial impairments using a mouse model. Oncotarget 2020; 11:3712-3722. [PMID: 33110478 PMCID: PMC7566807 DOI: 10.18632/oncotarget.27759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/10/2020] [Indexed: 01/21/2023] Open
Abstract
Myocardial damage in cancer patients is emphasized as a cause of death; however, there are not many murine cachexia models to evaluate cancer-derived heart disorder. Using the mouse cachexia model that we established previously, we investigated myocardial damage in tumor-bearing mice. In cachexic mice, decreased heart weight and myocardial volume, and dilated left ventricular lumen, and atrophied cardiomyocytes were noted. The cardiomyocytes also showed accumulated 8-hydroxydeoxyguanosine, decreased leucine zipper and EF-hand-containing transmembrane protein-1, and increased microtubule-associated protein light chain3-II. Levels of tumor necrosis factor-α and high-mobility group box-1 proteins in the myocardium were increased, and nuclear factor κB, a signaling molecule associated with these proteins, was activated. When rat cardiomyoblasts (H9c2 cells) were treated with mouse cachexia model ascites and subjected to flux analysis, both oxidative phosphorylation and glycolysis were suppressed, and the cells were in a quiescent state. These results are in good agreement with those previously reported on cancerous myocardial damage. The established mouse cachexia model can therefore be considered useful for analyzing cancer-derived myocardial damage.
Collapse
Affiliation(s)
- Yoshihiro Miyagawa
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Shota Nukaga
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan.,Division of Rehabilitation, Hanna Central Hospital, Ikoma, Nara 630-0243, Japan
| | - Takuya Mori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kei Goto
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan.,Division of Rehabilitation, Hoshida Minami Hospital, Katano, Osaka 576-0022, Japan
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Chie Nakashima
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Isao Kawahara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan.,Division of Rehabilitation, Hanna Central Hospital, Ikoma, Nara 630-0243, Japan
| | - Yi Luo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| |
Collapse
|
53
|
Nukaga S, Mori T, Miyagawa Y, Fujiwara-Tani R, Sasaki T, Fujii K, Mori S, Goto K, Kishi S, Nakashima C, Ohmori H, Kawahara I, Luo Y, Kuniyasu H. Combined administration of lauric acid and glucose improved cancer-derived cardiac atrophy in a mouse cachexia model. Cancer Sci 2020; 111:4605-4615. [PMID: 32969559 PMCID: PMC7734008 DOI: 10.1111/cas.14656] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/30/2022] Open
Abstract
Cancer‐derived myocardial damage is an important cause of death in cancer patients. However, the development of dietary interventions for treating such damage has not been advanced. Here, we investigated the effect of dietary intervention with lauric acid (LAA) and glucose, which was effective against skeletal muscle sarcopenia in a mouse cachexia model, on myocardial damage. Treatment of H9c2 rat cardiomyoblasts with lauric acid promoted mitochondrial respiration and increased ATP production by Seahorse flux analysis, but did not increase oxidative stress. Glycolysis was also promoted by LAA. In contrast, mitochondrial respiration and ATP production were suppressed, and oxidative stress was increased in an in vitro cachexia model in which cardiomyoblasts were treated with mouse cachexia ascites. Ascites‐treated H9c2 cells with concurrent treatment with LAA and high glucose showed that mitochondrial respiration and glycolysis were promoted more than that of the control, and ATP was restored to the level of the control. Oxidative stress was also reduced by the combined treatment. In the mouse cachexia model, myocardiac atrophy and decreased levels of a marker of muscle maturity, SDS‐soluble MYL1, were observed. When LAA in CE‐2 diet was orally administered alone, no significant rescue was observed in the cancer‐derived myocardial disorder. In contrast, combined oral administration of LAA and glucose recovered myocardial atrophy and MYL1 to levels observed in the control without increase in the cancer weight. Therefore, it is suggested that dietary intervention using a combination of LAA and glucose for cancer cachexia might improve cancer‐derived myocardial damage.
Collapse
Affiliation(s)
- Shota Nukaga
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan.,Division of Rehabilitation, Hanna Central Hospital, Ikoma, Japan
| | - Takuya Mori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Yoshihiro Miyagawa
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Kei Goto
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan.,Division of Rehabilitation, Hoshida Minami Hospital, Katano, Japan
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Chie Nakashima
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Isao Kawahara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan.,Division of Rehabilitation, Hanna Central Hospital, Ikoma, Japan
| | - Yi Luo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| |
Collapse
|
54
|
Rassaf T, Totzeck M, Backs J, Bokemeyer C, Hallek M, Hilfiker-Kleiner D, Hochhaus A, Lüftner D, Müller OJ, Neudorf U, Pfister R, von Haehling S, Lehmann LH, Bauersachs J. Onco-Cardiology: Consensus Paper of the German Cardiac Society, the German Society for Pediatric Cardiology and Congenital Heart Defects and the German Society for Hematology and Medical Oncology. Clin Res Cardiol 2020; 109:1197-1222. [PMID: 32405737 PMCID: PMC7515958 DOI: 10.1007/s00392-020-01636-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022]
Abstract
The acute and long-lasting side effects of modern multimodal tumour therapy significantly impair quality of life and survival of patients afflicted with malignancies. The key components of this therapy include radiotherapy, conventional chemotherapy, immunotherapy and targeted therapies. In addition to established tumour therapy strategies, up to 30 new therapies are approved each year with only incompletely characterised side effects. This consensus paper discusses the risk factors that contribute to the development of a potentially adverse reaction to tumour therapy and, in addition, defines specific side effect profiles for different treatment groups. The focus is on novel therapeutics and recommendations for the surveillance and treatment of specific patient groups.
Collapse
Affiliation(s)
- Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre Essen, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre Essen, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Johannes Backs
- Institute for Experimental Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with the Section Pneumology, Centre for Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany
| | | | - Andreas Hochhaus
- Department of Hematology and Medical Oncology, University Hospital Jena, Jena, Germany
| | - Diana Lüftner
- Department of Haematology, Oncology and Tumour Immunology, Charité, Humboldt University Berlin, Berlin, Germany
| | - Oliver J Müller
- Department of Internal Medicine III (Cardiology, Angiology and Internal Intensive Care Medicine), University Hospital Schleswig-Holstein, University of Kiel, Kiel, Germany
| | - Ulrich Neudorf
- Department of Pediatrics III, West German Heart and Vascular Centre Essen, University Hospital Essen, Essen, Germany
| | - Roman Pfister
- Clinic III for Internal Medicine, General and Interventional Cardiology, Electrophysiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Hospital Cologne, Cologne, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart Center Göttingen, University of Göttingen Medical Center and German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Lorenz H Lehmann
- Department of Cardiology, Angiology, Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
55
|
Kelm NQ, Straughn AR, Kakar SS. Withaferin A attenuates ovarian cancer-induced cardiac cachexia. PLoS One 2020; 15:e0236680. [PMID: 32722688 PMCID: PMC7386592 DOI: 10.1371/journal.pone.0236680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022] Open
Abstract
Cachexia is a common multifactorial syndrome in the advanced stages of cancer and accounts for approximately 20–30% of all cancer-related fatalities. In addition to the progressive loss of skeletal muscle mass, cancer results in impairments in cardiac function. We recently demonstrated that WFA attenuates the cachectic skeletal muscle phenotype induced by ovarian cancer. The purpose of this study was to investigate whether ovarian cancer induces cardiac cachexia, the possible pathway involved, and whether WFA attenuates cardiac cachexia. Xenografting of ovarian cancer induced cardiac cachexia, leading to the loss of normal heart functions. Treatment with WFA rescued the heart weight. Further, ovarian cancer induced systolic dysfunction and diastolic dysfunction Treatment with WFA preserved systolic function in tumor-bearing mice, but diastolic dysfunction was partially improved. In addition, WFA abrogated the ovarian cancer-induced reduction in cardiomyocyte cross-sectional area. Finally, treatment with WFA ameliorated fibrotic deposition in the hearts of tumor-bearing animals. We observed a tumor-induced MHC isoform switching from the adult MHCα to the embryonic MHCβ isoform, which was prevented by WFA treatment. Circulating Ang II level was increased significantly in the tumor-bearing, which was lowered by WFA treatment. Our results clearly demonstrated the induction of cardiac cachexia in response to ovarian tumors in female NSG mice. Further, we observed induction of proinflammatory markers through the AT1R pathway, which was ameliorated by WFA, in addition to amelioration of the cachectic phenotype, suggesting WFA as a potential therapeutic agent for cardiac cachexia in oncological paradigms.
Collapse
Affiliation(s)
- Natia Q. Kelm
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States of America
| | - Alex R. Straughn
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States of America
| | - Sham S. Kakar
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States of America
- Department of Physiology, University of Louisville, Louisville, KY, United States of America
- * E-mail:
| |
Collapse
|
56
|
Biswas AK, Acharyya S. Cancer-Associated Cachexia: A Systemic Consequence of Cancer Progression. ANNUAL REVIEW OF CANCER BIOLOGY 2020; 4:391-411. [DOI: 10.1146/annurev-cancerbio-030419-033642] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Cancer is a life-threatening disease that has plagued humans for centuries. The vast majority of cancer-related mortality results from metastasis. Indeed, the invasive growth of metastatic cancer cells in vital organs causes fatal organ dysfunction, but metastasis-related deaths also result from cachexia, a debilitating wasting syndrome characterized by an involuntary loss of skeletal muscle mass and function. In fact, about 80% of metastatic cancer patients suffer from cachexia, which often renders them too weak to tolerate standard doses of anticancer therapies and makes them susceptible to death from cardiac and respiratory failure. The goals of this review are to highlight important findings that help explain how cancer-induced systemic changes drive the development of cachexia and to discuss unmet challenges and potential therapeutic strategies targeting cachexia to improve the quality of life and survival of cancer patients.
Collapse
Affiliation(s)
- Anup K. Biswas
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Swarnali Acharyya
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
57
|
Evaluation of the Cardiac Protection Conferred by Proanthocyanidins in Grape Seeds against Development of Ehrlich Solid Tumors in Mice. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3530296. [PMID: 32016114 PMCID: PMC6985929 DOI: 10.1155/2020/3530296] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/10/2019] [Accepted: 12/21/2019] [Indexed: 02/08/2023]
Abstract
Examination of the antineoplastic effects of a range of chemical compounds is often undertaken via the transplantable tumor model of Ehrlich solid tumor (EST), which is a simulation of breast cancer. The purpose of this study was to explore how cardiac toxicity, damage, oxidative stress, and changes in the expressions of TNFα and apoptotic P53 triggered by EST could be countered with grape seed proanthocyanidins (GSPE). To that end, 50 female mice were used, with arbitrary and equal distribution into five groups, namely, the control group (G1), GSPE group (G2), EST group (G3), GSPE + EST (G4; cotreatment consisted of mice that received GSPE treatment at the beginning of EST induction over a period of 14 days), and EST + GSPE (G5; posttreatment consisted of mice with EST that received GSPE treatment for 14 days following the 14 days since the induction of EST). By comparison with the control group, the EST group had significantly higher levels of serum lactate dehydrogenase (LDH), creatine phosphokinase (CPK), creatine kinase MB (CK-MB), myoglobin, cardiac TBARS, nitric oxide (NO), total thiol and hydrogen peroxide, cardiac damage, and expression of P53 and TNFα. On the other hand, the EST group had significantly lower levels of cardiac catalase and total antioxidant (TAC) than the control group. Furthermore, better improvement in cardiac toxicity, oxidative stress, damage, apoptosis, and TNFα expressions was displayed by the cotreated (GSPE + EST) group than by the posttreated (EST + GSPE) group. This led to the conclusion that GSPE conferred cardiac protective and antioxidant effects against EST. This finding calls for more investigation on the benefits of grape seeds as adjuvant agents to prevent and treat cardiac toxicity.
Collapse
|
58
|
Obrezan AG, Shcherbakova NV. [Pathogenetic mechanisms of development of myocardial pathology in patients with malignant tumors: the current state of the problem]. ACTA ACUST UNITED AC 2020; 60:142-154. [PMID: 32345210 DOI: 10.18087/cardio.2020.2.n985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 12/23/2019] [Indexed: 11/18/2022]
Abstract
The myocardium, which has a high metabolic activity, responds to metabolic disorders and energy imbalance induced by a growing malignant tumor. In addition, the tumor itself can produce substances that directly affect metabolic processes and the life cycle of cells not involved in the neoplastic process, including cardiomyocytes. This review summarized and systematized current data on individual aspects of detrimental effects of oncogenes and tumor-related factors on the heart muscle and morpho-functional changes in the cardiovascular system of oncology patients. Also, the authors described in detail development of these pathogenetic mechanisms.
Collapse
Affiliation(s)
- A G Obrezan
- St. Petersburg State University, SOGAZ International Medical Center, St. Petersburg
| | | |
Collapse
|
59
|
Anthracycline Therapy Is Associated With Cardiomyocyte Atrophy and Preclinical Manifestations of Heart Disease. JACC Cardiovasc Imaging 2019; 11:1045-1055. [PMID: 30092965 DOI: 10.1016/j.jcmg.2018.05.012] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/05/2018] [Accepted: 05/11/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVES The goal of this study was to demonstrate that cardiac magnetic resonance could reveal anthracycline-induced early tissue remodeling and its relation to cardiac dysfunction and left ventricular (LV) atrophy. BACKGROUND Serum biomarkers of cardiac dysfunction, although elevated after chemotherapy, lack specificity for the mechanism of myocardial tissue alterations. METHODS A total of 27 women with breast cancer (mean age 51.8 ± 8.9 years, mean body mass index 26.9 ± 3.6 kg/m2), underwent cardiac magnetic resonance before and up to 3 times after anthracycline therapy. Cardiac magnetic resonance variables were LV ejection fraction, normalized T2-weighted signal intensity for myocardial edema, extracellular volume (ECV), LV cardiomyocyte mass, intracellular water lifetime (τic; a marker of cardiomyocyte size), and late gadolinium enhancement. RESULTS At baseline, patients had a relatively low (10-year) Framingham cardiovascular event risk (median 5%), normal LV ejection fractions (mean 69.4 ± 3.6%), and normal LV mass index (51.4 ± 8.0 g/m2), a mean ECV of 0.32 ± 0.038, mean τic of 169 ± 69 ms, and no late gadolinium enhancement. At 351 to 700 days after anthracycline therapy (240 mg/m2), mean LV ejection fraction had declined by 12% to 58 ± 6% (p < 0.001) and mean LV mass index by 19 g/m2 to 36 ± 6 g/m2 (p < 0.001), and mean ECV had increased by 0.037 to 0.36 ± 0.04 (p = 0.004), while mean τic had decreased by 62 ms to 119 ± 54 ms (p = 0.004). Myocardial edema peaked at about 146 to 231 days (p < 0.001). LV mass index was associated with τic (β = 4.1 ± 1.5 g/m2 per 100-ms increase in τic, p = 0.007) but not with ECV. Cardiac troponin T (mean 4.6 ± 1.4 pg/ml at baseline) increased significantly after anthracycline treatment (p < 0.001). Total LV cardiomyocyte mass, estimated as: (1 - ECV) × LV mass, declined more rapidly after anthracycline therapy, with peak cardiac troponin T >10 pg/ml. There was no evidence for any significant interaction between 10-year cardiovascular event risk and the effect of anthracycline therapy. CONCLUSIONS A decrease in LV mass after anthracycline therapy may result from cardiomyocyte atrophy, demonstrating that mechanisms other than interstitial fibrosis and edema can raise ECV. The loss of LV cardiomyocyte mass increased with the degree of cardiomyocyte injury, assessed by peak cardiac troponin T after anthracycline treatment. (Doxorubicin-Associated Cardiac Remodeling Followed by CMR in Breast Cancer Patients; NCT03000036).
Collapse
|
60
|
Kazemi-Bajestani SMR, Becher H, Butts C, Basappa NS, Smylie M, Joy AA, Sangha R, Gallivan A, Kavsak P, Chu Q, Baracos VE. Rapid atrophy of cardiac left ventricular mass in patients with non-small cell carcinoma of the lung. J Cachexia Sarcopenia Muscle 2019; 10:1070-1082. [PMID: 31293070 PMCID: PMC6818459 DOI: 10.1002/jcsm.12451] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 03/23/2019] [Accepted: 04/29/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Cancer is a systemic catabolic condition affecting skeletal muscle and fat. We aimed to determine whether cardiac atrophy occurs in this condition and assess its association with cardiac function, symptoms, and clinical outcomes. METHODS Treatment naïve metastatic non-small cell lung cancer patients (n = 50) were assessed prior to and 4 months after commencement of carboplatin-based palliative chemotherapy. Methods included echocardiography for left ventricular mass (LVM) and LV function [LV ejection fraction, global longitudinal strain (GLS), diastolic function], computed tomography to quantify skeletal muscle and total adipose tissue, Eastern Cooperative Oncology Group Performance Status (ECOG-PS), validated questionnaires for dyspnoea and fatigue, plasma biomarkers, tumour response to therapy, and overall survival. RESULTS During 112 ± 6 days, the median change in LVM was -8.9% [95% confidence interval (95% CI) -10.8 to -4.8, P < 0.001]. Quartiles of LVM loss were -20.1%, -12.9%, -4.8%, and +5.5%. Losses of muscle, adipose tissue, and LVM were frequently concurrent; LVM loss > median value was associated with loss of skeletal muscle [odds ratio (OR) = 4.5, 95% CI: 1.4-14.8, P=0.01] and loss of total adipose tissue (OR = 10.0, 95% CI: 2.7-36.7, P < 0.001). LVM loss was associated with decreased GLS (OR = 6.6, 95% CI: 1.9-22.7, P=0.003) but not with LV ejection fraction or diastolic function. In the population overall, plasma levels of C-reactive protein (P=0.008), high sensitivity troponin T (hs-TnT) (P=0.03), and galectin-3 (P=0.02) increased over time, while N-terminal pro B-type natriuretic peptide and hs-cTnI did not change over time. C-reactive protein was the only biomarker associated with LVM loss but at the univariate level only. Independent predictors of LVM loss were prior weight loss (adjusted OR = 10.2, 95% CI: 2.2-46.9, P=0.003) and tumour progression (adjusted OR = 14.6, 95% CI: 1.4-153.9, P=0.02). LVM loss was associated with exacerbations of fatigue (OR = 6.6, 95% CI: 1.9-22.7, P=0.003), dyspnoea (OR = 9.3, 95% CI: 2.4-35.8, P<0.001), and deterioration of performance status (OR = 4.8, 95% CI: 1.3-18.3,P=0.02). Patients with concurrent loss of LVM, skeletal muscle, and fat were more likely to deteriorate in all three symptom domains and to have reduced survival (P=0.05). CONCLUSIONS Intense LVM atrophy is associated with non-small cell lung cancer-induced cachexia. Loss of LVM was associated with emerging alterations of GLS, indicating subtle changes in left ventricular function. Longer term studies are needed to assess the full scope of cardiac atrophy and its impact. LVM atrophy arises in conjunction with losses of fat and skeletal muscle and is temporally associated with meaningful declines in performance status, worsening of fatigue, and dyspnoea, as well as poorer tumour response and decreased survival. The specific contribution of LVM atrophy to these outcomes requires further study.
Collapse
Affiliation(s)
| | - Harald Becher
- Department of Medicine, Division of Cardiology, Alberta Cardiovascular and Stroke Research Centre, Edmonton, Canada
| | - Charles Butts
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Naveen S Basappa
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Michael Smylie
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Anil Abraham Joy
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Randeep Sangha
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Andrea Gallivan
- Department of Oncology, Division of Palliative Care Medicine, University of Alberta, Edmonton, Canada
| | - Peter Kavsak
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Quincy Chu
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Vickie E Baracos
- Department of Oncology, Division of Palliative Care Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
61
|
Meijers WC, Maglione M, Bakker SJL, Oberhuber R, Kieneker LM, de Jong S, Haubner BJ, Nagengast WB, Lyon AR, van der Vegt B, van Veldhuisen DJ, Westenbrink BD, van der Meer P, Silljé HHW, de Boer RA. Heart Failure Stimulates Tumor Growth by Circulating Factors. Circulation 2019; 138:678-691. [PMID: 29459363 DOI: 10.1161/circulationaha.117.030816] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Heart failure (HF) survival has improved, and nowadays, many patients with HF die of noncardiac causes, including cancer. Our aim was to investigate whether a causal relationship exists between HF and the development of cancer. METHODS HF was induced by inflicting large anterior myocardial infarction in APCmin mice, which are prone to developing precancerous intestinal tumors, and tumor growth was measured. In addition, to rule out hemodynamic impairment, a heterotopic heart transplantation model was used in which an infarcted or sham-operated heart was transplanted into a recipient mouse while the native heart was left in situ. After 6 weeks, tumor number, volume, and proliferation were quantified. Candidate secreted proteins were selected because they were previously associated both with (colon) tumor growth and with myocardial production in post-myocardial infarction proteomic studies. Myocardial gene expression levels of these selected candidates were analyzed, as well as their proliferative effects on HT-29 (colon cancer) cells. We validated these candidates by measuring them in plasma of healthy subjects and patients with HF. Finally, we associated the relation between cardiac specific and inflammatory biomarkers and new-onset cancer in a large, prospective general population cohort. RESULTS The presence of failing hearts, both native and heterotopically transplanted, resulted in significantly increased intestinal tumor load of 2.4-fold in APCmin mice (all P<0.0001). The severity of left ventricular dysfunction and fibrotic scar strongly correlated with tumor growth ( P=0.002 and P=0.016, respectively). We identified several proteins (including serpinA3 and A1, fibronectin, ceruloplasmin, and paraoxonase 1) that were elevated in human patients with chronic HF (n=101) compared with healthy subjects (n=180; P<0.001). Functionally, serpinA3 resulted in marked proliferation effects in human colon cancer (HT-29) cells, associated with Akt-S6 phosphorylation. Finally, elevated cardiac and inflammation biomarkers in apparently healthy humans (n=8319) were predictive of new-onset cancer (n=1124) independently of risk factors for cancer (age, smoking status, and body mass index). CONCLUSIONS We demonstrate that the presence of HF is associated with enhanced tumor growth and that this is independent of hemodynamic impairment and could be caused by cardiac excreted factors. A diagnosis of HF may therefore be considered a risk factor for incident cancer.
Collapse
Affiliation(s)
- Wouter C Meijers
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Manuel Maglione
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery (M.M., R.O.)
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology (S.J.L.B., L.M.K.), University Medical Center Groningen, University of Groningen, The Netherlands
| | - Rupert Oberhuber
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery (M.M., R.O.)
| | - Lyanne M Kieneker
- Department of Internal Medicine, Division of Nephrology (S.J.L.B., L.M.K.), University Medical Center Groningen, University of Groningen, The Netherlands
| | | | - Bernhard J Haubner
- Department of Internal Medicine III (Cardiology and Angiology) (B.J.H.), Medical University of Innsbruck, Austria
| | | | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital, United Kingdom (A.R.L.)
| | | | | | - B Daan Westenbrink
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Peter van der Meer
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Herman H W Silljé
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| | - Rudolf A de Boer
- Department of Cardiology (W.C.M., D.J.v.V., B.D.W., P.v.d.M., H.H.W.S., R.A.d.B.)
| |
Collapse
|
62
|
Scott JM, Dillon EL, Kinsky M, Chamberlain A, McCammon S, Jupiter D, Willis M, Hatch S, Richardson G, Danesi C, Randolph K, Durham W, Wright T, Urban R, Sheffield-Moore M. Effects of adjunct testosterone on cardiac morphology and function in advanced cancers: an ancillary analysis of a randomized controlled trial. BMC Cancer 2019; 19:778. [PMID: 31391011 PMCID: PMC6686390 DOI: 10.1186/s12885-019-6006-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Adjunct testosterone therapy improves lean body mass, quality of life, and physical activity in patients with advanced cancers; however, the effects of testosterone on cardiac morphology and function are unknown. Accordingly, as an ancillary analysis of a randomized, placebo-controlled trial investigating the efficacy of testosterone supplementation on body composition in men and women with advanced cancers, we explored whether testosterone supplementation could prevent or reverse left ventricular (LV) atrophy and dysfunction. METHODS Men and women recently diagnosed with late stage (≥IIB) or recurrent head and neck or cervical cancer who were scheduled to receive standard of care chemotherapy or concurrent chemoradiation were administered an adjunct 7 week treatment of weekly intramuscular injections of either 100 mg testosterone (T, n = 1 M/5F) or placebo (P, n = 6 M/4F) in a double-blinded randomized fashion. LV morphology (wall thickness), systolic function (ejection fraction, EF), diastolic function (E/A; E'/E), arterial elastance (Ea), end-systolic elastance (Ees), and ventricular-arterial coupling (Ea/Ees) were assessed. RESULTS No significant differences were observed in LV posterior wall thickness in placebo (pre: 1.10 ± 0.1 cm; post: 1.16 ± 0.2 cm; p = 0.11) or testosterone groups (pre: 0.99 ± 0.1 cm; post: 1.14 ± 0.20 cm; p = 0.22). Compared with placebo, testosterone significantly improved LVEF (placebo: - 1.8 ± 4.3%; testosterone: + 6.2 ± 4.3%; p < 0.05), Ea (placebo: 0.0 ± 0.2 mmHg/mL; testosterone: - 0.3 ± 0.2 mmHg/mL; p < 0.05), and Ea/Ees (placebo: 0.0 ± 0.1; testosterone: - 0.2 ± 0.1; p < 0.05). CONCLUSIONS In patients with advanced cancers, testosterone was associated with favorable changes in left ventricular systolic function, arterial elastance, and ventricular-arterial coupling. Given the small sample size, the promising multisystem benefits of testosterone warrants further evaluation in a definitive randomized trial. TRIAL REGISTRATION This study was prospectively registered on ClinicalTrials.gov (NCT00878995; date of registration: April 9, 2009).
Collapse
Affiliation(s)
- Jessica M Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Lichar Dillon
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Michael Kinsky
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Albert Chamberlain
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Susan McCammon
- Department of Otolaryngology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel Jupiter
- Department of Preventive Medicine and Community Health, The University of Texas Medical Branch, Galveston, TX, USA
| | - Maurice Willis
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Sandra Hatch
- Department of Radiation Oncology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Gwyn Richardson
- Department of Gynecologic Oncology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Christopher Danesi
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Kathleen Randolph
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
- Department of Health and Kinesiology, Texas A&M University, 155 Ireland St., College Station, TX, TX 77845, USA
| | - William Durham
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Traver Wright
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
- Department of Health and Kinesiology, Texas A&M University, 155 Ireland St., College Station, TX, TX 77845, USA
| | - Randall Urban
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Melinda Sheffield-Moore
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA.
- Department of Health and Kinesiology, Texas A&M University, 155 Ireland St., College Station, TX, TX 77845, USA.
| |
Collapse
|
63
|
Brancaccio M, Pirozzi F, Hirsch E, Ghigo A. Mechanisms underlying the cross-talk between heart and cancer. J Physiol 2019; 598:3015-3027. [PMID: 31278748 DOI: 10.1113/jp276746] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases and cancer remain the leading cause of death worldwide. Despite the fact that these two conditions have long been considered as distinct clinical entities, recent epidemiological and experimental studies suggest that they should be contemplated and treated as co-morbidities. Heart failure represents nowadays a well-established complication of cancer, primarily as a consequence of the aggressive use of cardiotoxic anti-cancer treatments. On the other hand, the provocative idea that heart failure can prime carcinogenesis has started to emerge, though the molecular basis is still to be fully elucidated. This review summarizes the current knowledge on the mechanisms underlying the bidirectional communication between the failing heart and the cancer. We will discuss and/or speculate on the role of molecular mediators released by either the tumour or the heart that can potentially link heart failure and cancer.
Collapse
Affiliation(s)
- Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
64
|
Abstract
Cancer has been shown to negatively stimulate autophagy, leading to a decline in cardiac function. Although exercise is cardioprotective, its influence over autophagy-mediated tumor growth and cardiac function are not well defined. PURPOSE This study aimed to determine the effect of exercise on tumor morphology and cardiac function. METHODS Fisher 344 rats (n = 28) were assigned to one of four groups: 1) sedentary non-tumor bearing (SED), 2) sedentary tumor bearing (SED + T), 3) wheel run non-tumor bearing (WR), or 4) wheel run tumor bearing (WR + T). Rats remained sedentary or exercised for 6 wk. At week 4, rats in tumor groups were inoculated with MatBIII tumor cells. At week 6, cardiac function was measured. RESULTS SED + T animals exhibited significantly lower left ventricular developed pressure when compared with SED, WR, and WR + T (P < 0.05). This coincided with a significant increase in cardiac autophagy (increased LC3-II) in SED + T animals when compared with SED, WR, and WR + T (P < 0.05). Furthermore, SED + T hearts showed a significant increase in β-myosin heavy chain expression versus nontumor groups (P < 0.05). Tumor mass was significantly larger (P < 0.001) in SED + T animals when compared with WR + T animals, which was accompanied by a significant increase in tumor LC3-II protein expression (P < 0.05). CONCLUSION Nonexercised tumor-bearing rats showed severe cardiac dysfunction and excessive, maladaptive autophagy in the heart and tumors. Voluntary exercise preserved cardiac function and attenuated the autophagic response in heart and tumor tissues. This preservation may be related to the reduced tumor growth in aerobically exercised rats, to the improved regulation of autophagy by exercise, or both.
Collapse
Affiliation(s)
- Traci L Parry
- McAllister Heart Institute and Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC
| | - Reid Hayward
- School of Sport and Exercise Science, and the University of Northern Colorado Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO
| |
Collapse
|
65
|
Baumfalk DR, Opoku-Acheampong AB, Caldwell JT, Ade CJ, Copp SW, Musch TI, Behnke BJ. Effects of prostate cancer and exercise training on left ventricular function and cardiac and skeletal muscle mass. J Appl Physiol (1985) 2019; 126:668-680. [PMID: 30571286 DOI: 10.1152/japplphysiol.00829.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Prostate cancer was found to reduce cardiac and left ventricle (LV) masses in association with diminished exercise capacity in rats. We tested the hypothesis that exercise training will mitigate prostate cancer-induced cardiac and skeletal muscle atrophy and improve LV function versus sedentary tumor-bearing counterparts. Copenhagen rats ( n = 39; ~5 mo old) were randomized into four groups: exercise-trained tumor-bearing (EXTB) or control (EXCON) and sedentary tumor-bearing (SEDTB) or control (SEDCON). Dunning R-3327 prostate cancer cells were injected orthotopically in 19 of the 39 animals. Treadmill exercise training was performed for 60 min/day for ~30 days. Animals underwent echocardiography to examine ventricle dimensions "Pre-" cancer injection or exercise (PRE) and 15 (Post 1) and 32-35 (Post 2) days after cancer cell injection with tissues collected after Post 2. LV TNF-α and IL-6 concentrations were measured post mortem. Cardiac and LV mass of SEDTB animals were lower than all groups ( P < 0.05). Tumor mass was negatively correlated with LV mass in EXTB (-0.75, P < 0.02) and SEDTB animals (-0.72, P < 0.02). EXCON group had higher stroke volume Post 2 assessment compared with both sedentary groups ( P < 0.05) but not EXTB animals. No difference in LV [IL-6] or [TNF-α] was found between the cancer groups. The current investigation demonstrates prostate cancer, independent of anticancer treatment, significantly reduces cardiac mass and LV mass as well as locomotor muscle masses. However, moderate-intensity exercise training can mitigate cardiac and skeletal muscle atrophy with prostate cancer and preserve the cardiac phenotype (i.e., mass and function) to that of the healthy sedentary group. NEW & NOTEWORTHY This study demonstrates the atrophic effects of prostate cancer on cardiac and skeletal muscle mass independent of anticancer treatment(s) that can be mitigated with moderate-intensity exercise. These findings have important implications for potentially improving the quality of life as well as therapeutic outcomes for patients with prostate cancer.
Collapse
Affiliation(s)
- Dryden R Baumfalk
- Department of Kinesiology, Kansas State University , Manhattan, Kansas
| | | | - Jacob T Caldwell
- Department of Kinesiology, Kansas State University , Manhattan, Kansas
| | - Carl J Ade
- Department of Kinesiology, Kansas State University , Manhattan, Kansas.,Johnson Cancer Research Center, Kansas State University , Manhattan, Kansas
| | - Steven W Copp
- Department of Kinesiology, Kansas State University , Manhattan, Kansas
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University , Manhattan, Kansas.,Department of Anatomy and Physiology, Kansas State University , Manhattan, Kansas
| | - Bradley J Behnke
- Department of Kinesiology, Kansas State University , Manhattan, Kansas.,Johnson Cancer Research Center, Kansas State University , Manhattan, Kansas
| |
Collapse
|
66
|
Willis MS, Parry TL, Brown DI, Mota RI, Huang W, Beak JY, Sola M, Zhou C, Hicks ST, Caughey MC, D’agostino RB, Jordan J, Hundley WG, Jensen BC. Doxorubicin Exposure Causes Subacute Cardiac Atrophy Dependent on the Striated Muscle-Specific Ubiquitin Ligase MuRF1. Circ Heart Fail 2019; 12:e005234. [PMID: 30871347 PMCID: PMC6422170 DOI: 10.1161/circheartfailure.118.005234] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 01/17/2019] [Indexed: 01/30/2023]
Abstract
Background Anthracycline chemotherapeutics, such as doxorubicin, are used widely in the treatment of numerous malignancies. The primary dose-limiting adverse effect of anthracyclines is cardiotoxicity that often presents as heart failure due to dilated cardiomyopathy years after anthracycline exposure. Recent data from animal studies indicate that anthracyclines cause cardiac atrophy. The timing of onset and underlying mechanisms are not well defined, and the relevance of these findings to human disease is unclear. Methods and Results Wild-type mice were sacrificed 1 week after intraperitoneal administration of doxorubicin (1-25 mg/kg), revealing a dose-dependent decrease in cardiac mass ( R2=0.64; P<0.0001) and a significant decrease in cardiomyocyte cross-sectional area (336±29 versus 188±14 µm2; P<0.0001). Myocardial tissue analysis identified a dose-dependent upregulation of the ubiquitin ligase, MuRF1 (muscle ring finger-1; R2=0.91; P=0.003) and a molecular profile of muscle atrophy. To investigate the determinants of doxorubicin-induced cardiac atrophy, we administered doxorubicin 20 mg/kg to mice lacking MuRF1 (MuRF1-/-) and wild-type littermates. MuRF1-/- mice were protected from cardiac atrophy and exhibited no reduction in contractile function. To explore the clinical relevance of these findings, we analyzed cardiac magnetic resonance imaging data from 70 patients in the DETECT-1 cohort and found that anthracycline exposure was associated with decreased cardiac mass evident within 1 month and persisting to 6 months after initiation. Conclusions Doxorubicin causes a subacute decrease in cardiac mass in both mice and humans. In mice, doxorubicin-induced cardiac atrophy is dependent on MuRF1. These findings suggest that therapies directed at preventing or reversing cardiac atrophy might preserve the cardiac function of cancer patients receiving anthracyclines.
Collapse
Affiliation(s)
- Monte S. Willis
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
- Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN
| | - Traci L. Parry
- Department of Pathology & Laboratory Medicine, University of North Carolina School of Medicine
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - David I. Brown
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - Roberto I. Mota
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - Wei Huang
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - Ju Youn Beak
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - Michael Sola
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - Cynthia Zhou
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - Sean T Hicks
- McAllister Heart Institute, University of North Carolina School of Medicine
| | - Melissa C. Caughey
- Department of Medicine, Division of Cardiology, University of North Carolina School of Medicine
| | | | - Jennifer Jordan
- Section on Cardiovascular Medicine, Wake Forest Health Sciences
| | | | - Brian C. Jensen
- McAllister Heart Institute, University of North Carolina School of Medicine
- Department of Pharmacology, University of North Carolina School of Medicine
- Department of Medicine, Division of Cardiology, University of North Carolina School of Medicine
| |
Collapse
|
67
|
Schmidt SF, Rohm M, Herzig S, Berriel Diaz M. Cancer Cachexia: More Than Skeletal Muscle Wasting. Trends Cancer 2018; 4:849-860. [DOI: 10.1016/j.trecan.2018.10.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/21/2022]
|
68
|
Antunes J, Ferreira RM, Moreira-Gonçalves D. Exercise Training as Therapy for Cancer-Induced Cardiac Cachexia. Trends Mol Med 2018; 24:709-727. [DOI: 10.1016/j.molmed.2018.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 12/27/2022]
|
69
|
mTOR and Tumor Cachexia. Int J Mol Sci 2018; 19:ijms19082225. [PMID: 30061533 PMCID: PMC6121479 DOI: 10.3390/ijms19082225] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022] Open
Abstract
Cancer cachexia affects most patients with advanced forms of cancers. It is mainly characterized by weight loss, due to muscle and adipose mass depletion. As cachexia is associated with increased morbidity and mortality in cancer patients, identifying the underlying mechanisms leading to cachexia is essential in order to design novel therapeutic strategies. The mechanistic target of rapamycin (mTOR) is a major intracellular signalling intermediary that participates in cell growth by upregulating anabolic processes such as protein and lipid synthesis. Accordingly, emerging evidence suggests that mTOR and mTOR inhibitors influence cancer cachexia. Here, we review the role of mTOR in cellular processes involved in cancer cachexia and highlight the studies supporting the contribution of mTOR in cancer cachexia.
Collapse
|
70
|
Nissinen TA, Hentilä J, Penna F, Lampinen A, Lautaoja JH, Fachada V, Holopainen T, Ritvos O, Kivelä R, Hulmi JJ. Treating cachexia using soluble ACVR2B improves survival, alters mTOR localization, and attenuates liver and spleen responses. J Cachexia Sarcopenia Muscle 2018; 9:514-529. [PMID: 29722201 PMCID: PMC5989872 DOI: 10.1002/jcsm.12310] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/20/2018] [Accepted: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cancer cachexia increases morbidity and mortality, and blocking of activin receptor ligands has improved survival in experimental cancer. However, the underlying mechanisms have not yet been fully uncovered. METHODS The effects of blocking activin receptor type 2 (ACVR2) ligands on both muscle and non-muscle tissues were investigated in a preclinical model of cancer cachexia using a recombinant soluble ACVR2B (sACVR2B-Fc). Treatment with sACVR2B-Fc was applied either only before the tumour formation or with continued treatment both before and after tumour formation. The potential roles of muscle and non-muscle tissues in cancer cachexia were investigated in order to understand the possible mechanisms of improved survival mediated by ACVR2 ligand blocking. RESULTS Blocking of ACVR2 ligands improved survival in tumour-bearing mice only when the mice were treated both before and after the tumour formation. This occurred without effects on tumour growth, production of pro-inflammatory cytokines or the level of physical activity. ACVR2 ligand blocking was associated with increased muscle (limb and diaphragm) mass and attenuation of both hepatic protein synthesis and splenomegaly. Especially, the effects on the liver and the spleen were observed independent of the treatment protocol. The prevention of splenomegaly by sACVR2B-Fc was not explained by decreased markers of myeloid-derived suppressor cells. Decreased tibialis anterior, diaphragm, and heart protein synthesis were observed in cachectic mice. This was associated with decreased mechanistic target of rapamycin (mTOR) colocalization with late-endosomes/lysosomes, which correlated with cachexia and reduced muscle protein synthesis. CONCLUSIONS The prolonged survival with continued ACVR2 ligand blocking could potentially be attributed in part to the maintenance of limb and respiratory muscle mass, but many observed non-muscle effects suggest that the effect may be more complex than previously thought. Our novel finding showing decreased mTOR localization in skeletal muscle with lysosomes/late-endosomes in cancer opens up new research questions and possible treatment options for cachexia.
Collapse
Affiliation(s)
- Tuuli A Nissinen
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Jaakko Hentilä
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello, Turin, 10125, Italy
| | - Anita Lampinen
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Juulia H Lautaoja
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Vasco Fachada
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Tanja Holopainen
- Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Riikka Kivelä
- Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Juha J Hulmi
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| |
Collapse
|
71
|
Nunes RAB, Bruno JDA, Ramirez HSM, Demarchi LMMF. Case 2/2018 - 73-Year-Old Male with Ischemic Cardiomyopathy, Cachexia and Shock. Arq Bras Cardiol 2018; 110:388-392. [PMID: 29791579 PMCID: PMC5941965 DOI: 10.5935/abc.20180065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
72
|
Subcutaneous Ehrlich Ascites Carcinoma mice model for studying cancer-induced cardiomyopathy. Sci Rep 2018; 8:5599. [PMID: 29618792 PMCID: PMC5884778 DOI: 10.1038/s41598-018-23669-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/08/2018] [Indexed: 02/06/2023] Open
Abstract
Cardiomyopathy is one of the characteristic features of cancer. In this study, we establish a suitable model to study breast cancer-induced cardiomyopathy in mice. We used Ehrlich Ascites Carcinoma cells to induce subcutaneous tumor in 129/SvJ mice and studied its effect on heart function. In Ehrlich Ascites Carcinoma bearing mice, we found significant reduction in left ventricle wall thickness, ejection fraction, and fractional shortening increase in left ventricle internal diameter. We found higher muscle atrophy, degeneration, fibrosis, expression of cell-adhesion molecules and cell death in tumor-bearing mice hearts. As observed in cancer patients, we found that mTOR, a key signalling molecule responsible for maintaining cell growth and autophagy was suppressed in this model. Tumor bearing mice hearts show increased expression and nuclear localization of TFEB and FoxO3a transcription factors, which are involved in the upregulation of muscle atrophy genes, lysosomal biogenesis genes and autophagy genes. We propose that Ehrlich Ascites Carcinoma induced tumor can be used as a model to identify potential therapeutic targets for the treatment of heart failure in patients suffering from cancer-induced cardiomyopathy. This model can also be used to test the adverse consequences of cancer chemotherapy in heart.
Collapse
|
73
|
Hulmi JJ, Nissinen TA, Räsänen M, Degerman J, Lautaoja JH, Hemanthakumar KA, Backman JT, Ritvos O, Silvennoinen M, Kivelä R. Prevention of chemotherapy-induced cachexia by ACVR2B ligand blocking has different effects on heart and skeletal muscle. J Cachexia Sarcopenia Muscle 2018; 9:417-432. [PMID: 29230965 PMCID: PMC5879968 DOI: 10.1002/jcsm.12265] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/15/2017] [Accepted: 10/12/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Toxicity of chemotherapy on skeletal muscles and the heart may significantly contribute to cancer cachexia, mortality, and decreased quality of life. Doxorubicin (DOX) is an effective cytostatic agent, which unfortunately has toxic effects on many healthy tissues. Blocking of activin receptor type IIB (ACVR2B) ligands is an often used strategy to prevent skeletal muscle loss, but its effects on the heart are relatively unknown. METHODS The effects of DOX treatment with or without pre-treatment with soluble ACVR2B-Fc (sACVR2B-Fc) were investigated. The mice were randomly assigned into one of the three groups: (1) vehicle (PBS)-treated controls, (2) DOX-treated mice (DOX), and (3) DOX-treated mice administered with sACVR2B-Fc during the experiment (DOX + sACVR2B-Fc). DOX was administered with a cumulative dose of 24 mg/kg during 2 weeks to investigate cachexia outcome in the heart and skeletal muscle. To understand similarities and differences between skeletal and cardiac muscles in their responses to chemotherapy, the tissues were collected 20 h after a single DOX (15 mg/kg) injection and analysed with genome-wide transcriptomics and mRNA and protein analyses. The combination group was pre-treated with sACVR2B-Fc 48 h before DOX administration. Major findings were also studied in mice receiving only sACVR2B-Fc. RESULTS The DOX treatment induced similar (~10%) wasting in skeletal muscle and the heart. However, transcriptional changes in response to DOX were much greater in skeletal muscle. Pathway analysis and unbiased transcription factor analysis showed that p53-p21-REDD1 is the main common pathway activated by DOX in both skeletal and cardiac muscles. These changes were attenuated by blocking ACVR2B ligands especially in skeletal muscle. Tceal7 (3-fold to 5-fold increase), transferrin receptor (1.5-fold increase), and Ccl21 (0.6-fold to 0.9-fold decrease) were identified as novel genes responsive to blocking ACVR2B ligands. Overall, at the transcriptome level, ACVR2B ligand blocking had only minor influence in the heart while it had marked effects in skeletal muscle. The same was also true for the effects on tissue wasting. This may be explained in part by about 18-fold higher gene expression of myostatin in skeletal muscle compared with the heart. CONCLUSIONS Cardiac and skeletal muscles display similar atrophy after DOX treatment, but the mechanisms for this may differ between the tissues. The present results suggest that p53-p21-REDD1 signalling is the main common DOX-activated pathway in these tissues and that blocking activin receptor ligands attenuates this response, especially in skeletal muscle supporting the overall stronger effects of this treatment in skeletal muscles.
Collapse
Affiliation(s)
- Juha J Hulmi
- Biology of Physical Activity, Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuuli A Nissinen
- Biology of Physical Activity, Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Markus Räsänen
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Joni Degerman
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juulia H Lautaoja
- Biology of Physical Activity, Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mika Silvennoinen
- Biology of Physical Activity, Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Riikka Kivelä
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
74
|
Tilemann LM, Heckmann MB, Katus HA, Lehmann LH, Müller OJ. Cardio-oncology: conflicting priorities of anticancer treatment and cardiovascular outcome. Clin Res Cardiol 2018; 107:271-280. [PMID: 29453595 PMCID: PMC5869944 DOI: 10.1007/s00392-018-1202-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/11/2018] [Indexed: 12/03/2022]
Abstract
BACKGROUND This article about the emerging field of cardio-oncology highlights typical side effects of oncological therapies in the cardiovascular system, cardiovascular complications of malignancies itself, and potential preventive or therapeutic modalities. METHODS We performed a selective literature search in PubMed until September 2016. RESULTS Cardiovascular events in cancer patients can be frequently attributed to oncological therapies or to the underlying malignancy itself. Furthermore, many patients with cancer have pre-existing cardiovascular diseases that can be aggravated by the malignancy or its therapy. Cardiovascular abnormalities in oncological patients comprise a broad spectrum from alterations in electrophysiological, laboratory or imaging tests to the occurrence of thromboembolic, ischemic or rhythmological events and the impairment of left ventricular function or manifest heart failure. DISCUSSION A close interdisciplinary collaboration between oncologists and cardiologists/angiologists as well as an increased awareness of potential cardiovascular complications could improve clinical care of cancer patients and provides a basis for an improved understanding of underlying mechanisms of cardiovascular morbidity.
Collapse
Affiliation(s)
- Lisa M Tilemann
- Abteilung für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Standort Heidelberg, Mannheim, Germany
| | - Markus B Heckmann
- Abteilung für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Standort Heidelberg, Mannheim, Germany
| | - Hugo A Katus
- Abteilung für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Standort Heidelberg, Mannheim, Germany
| | - Lorenz H Lehmann
- Abteilung für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Standort Heidelberg, Mannheim, Germany.
| | - Oliver J Müller
- Abteilung für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Standort Heidelberg, Mannheim, Germany.
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| |
Collapse
|
75
|
Padrão AI, Nogueira-Ferreira R, Vitorino R, Carvalho D, Correia C, Neuparth MJ, Pires MJ, Faustino-Rocha AI, Santos LL, Oliveira PA, Duarte JA, Moreira-Gonçalves D, Ferreira R. Exercise training protects against cancer-induced cardiac remodeling in an animal model of urothelial carcinoma. Arch Biochem Biophys 2018; 645:12-18. [PMID: 29548774 DOI: 10.1016/j.abb.2018.03.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/20/2018] [Accepted: 03/12/2018] [Indexed: 12/24/2022]
Abstract
Limiting cancer-induced cardiac damage has become an increasingly important issue to improve survival rates and quality of life. Exercise training has been shown to reduce cardiovascular complications in several diseases; however, its therapeutic role against cardiovascular consequences of cancer is in its infancy. In order to add new insights on the potential therapeutic effect of exercise training on cancer-related cardiac dysfunction, we used an animal model of urothelial carcinoma submitted to 13 weeks of treadmill exercise after 20 weeks of exposure to the carcinogenic N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN). Data showed that 13 weeks of treadmill exercise reverted cancer-induced cardiomyocytes atrophy and fibrosis, improved cardiac oxidative capacity given by citrate synthase activity and MnSOD content, and increased the levels of the mitochondrial biogenesis markers PGC-1α and mtTFA. Moreover, exercise training reverted cancer-induced decrease of cardiac c-kit levels suggesting enhanced regenerative ability of heart. These cardiac adaptations to exercise were related to a lower incidence of malignant urothelial lesions and less signs of inflammation. Taken together, data from the present study support the beneficial effect of exercise training when started after cancer diagnosis, envisioning the improvement of the cardiovascular function.
Collapse
Affiliation(s)
- Ana Isabel Padrão
- QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal; CIAFEL, Faculty of Sports, University of Porto, R. Dr. Plácido da Costa 91, 4200-450, Porto, Portugal
| | - Rita Nogueira-Ferreira
- iBiMED, Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal; Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal; Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Dulce Carvalho
- QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Catarina Correia
- QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Maria João Neuparth
- CIAFEL, Faculty of Sports, University of Porto, R. Dr. Plácido da Costa 91, 4200-450, Porto, Portugal
| | - Maria João Pires
- CITAB, Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5001-911 Vila Real, Portugal
| | - Ana Isabel Faustino-Rocha
- CITAB, Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5001-911 Vila Real, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center of Instituto Português de Oncologia, R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Paula Alexandra Oliveira
- CITAB, Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5001-911 Vila Real, Portugal
| | - José Alberto Duarte
- CIAFEL, Faculty of Sports, University of Porto, R. Dr. Plácido da Costa 91, 4200-450, Porto, Portugal
| | - Daniel Moreira-Gonçalves
- CIAFEL, Faculty of Sports, University of Porto, R. Dr. Plácido da Costa 91, 4200-450, Porto, Portugal; Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Rita Ferreira
- QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
76
|
Abstract
Cancer-associated cachexia is a disorder characterized by loss of body weight with specific losses of skeletal muscle and adipose tissue. Cachexia is driven by a variable combination of reduced food intake and metabolic changes, including elevated energy expenditure, excess catabolism and inflammation. Cachexia is highly associated with cancers of the pancreas, oesophagus, stomach, lung, liver and bowel; this group of malignancies is responsible for half of all cancer deaths worldwide. Cachexia involves diverse mediators derived from the cancer cells and cells within the tumour microenvironment, including inflammatory and immune cells. In addition, endocrine, metabolic and central nervous system perturbations combine with these mediators to elicit catabolic changes in skeletal and cardiac muscle and adipose tissue. At the tissue level, mechanisms include activation of inflammation, proteolysis, autophagy and lipolysis. Cachexia associates with a multitude of morbidities encompassing functional, metabolic and immune disorders as well as aggravated toxicity and complications of cancer therapy. Patients experience impaired quality of life, reduced physical, emotional and social well-being and increased use of healthcare resources. To date, no effective medical intervention completely reverses cachexia and there are no approved drug therapies. Adequate nutritional support remains a mainstay of cachexia therapy, whereas drugs that target overactivation of catabolic processes, cell injury and inflammation are currently under investigation.
Collapse
Affiliation(s)
- Vickie E Baracos
- Division of Palliative Care Medicine, Department of Oncology, University of Alberta, Cross Cancer Institute 11560 University Avenue, Edmonton, T6G 1Z2 Alberta, Canada
| | - Lisa Martin
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Murray Korc
- Section of Endocrinology, Departments of Medicine and Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Denis C Guttridge
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
| | - Kenneth C H Fearon
- Clinical and Surgical Sciences, School of Clinical Sciences and Community Health, Royal Infirmary, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
77
|
Zheng P, Li J, Kros JM. Breakthroughs in modern cancer therapy and elusive cardiotoxicity: Critical research-practice gaps, challenges, and insights. Med Res Rev 2018; 38:325-376. [PMID: 28862319 PMCID: PMC5763363 DOI: 10.1002/med.21463] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/16/2022]
Abstract
To date, five cancer treatment modalities have been defined. The three traditional modalities of cancer treatment are surgery, radiotherapy, and conventional chemotherapy, and the two modern modalities include molecularly targeted therapy (the fourth modality) and immunotherapy (the fifth modality). The cardiotoxicity associated with conventional chemotherapy and radiotherapy is well known. Similar adverse cardiac events are resurging with the fourth modality. Aside from the conventional and newer targeted agents, even the most newly developed, immune-based therapeutic modalities of anticancer treatment (the fifth modality), e.g., immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy, have unfortunately led to potentially lethal cardiotoxicity in patients. Cardiac complications represent unresolved and potentially life-threatening conditions in cancer survivors, while effective clinical management remains quite challenging. As a consequence, morbidity and mortality related to cardiac complications now threaten to offset some favorable benefits of modern cancer treatments in cancer-related survival, regardless of the oncologic prognosis. This review focuses on identifying critical research-practice gaps, addressing real-world challenges and pinpointing real-time insights in general terms under the context of clinical cardiotoxicity induced by the fourth and fifth modalities of cancer treatment. The information ranges from basic science to clinical management in the field of cardio-oncology and crosses the interface between oncology and onco-pharmacology. The complexity of the ongoing clinical problem is addressed at different levels. A better understanding of these research-practice gaps may advance research initiatives on the development of mechanism-based diagnoses and treatments for the effective clinical management of cardiotoxicity.
Collapse
Affiliation(s)
- Ping‐Pin Zheng
- Cardio‐Oncology Research GroupErasmus Medical CenterRotterdamthe Netherlands
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| | - Jin Li
- Department of OncologyShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Johan M Kros
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| |
Collapse
|
78
|
Barkhudaryan A, Scherbakov N, Springer J, Doehner W. Cardiac muscle wasting in individuals with cancer cachexia. ESC Heart Fail 2017; 4:458-467. [PMID: 29154433 PMCID: PMC5695173 DOI: 10.1002/ehf2.12184] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 03/28/2017] [Accepted: 05/01/2017] [Indexed: 12/25/2022] Open
Abstract
Aims Cachexia is a severe complication of cancer that adversely affects the course of the disease and is associated with high rates of mortality. Patients with cancer manifest symptoms, such as fatigue, shortness of breath, and impaired exercise tolerance, which are clinical signs of chronic heart failure. The aim of this study was to evaluate cardiac muscle wasting in cancer individuals. Methods and results We retrospectively analysed 177 individuals who died of cancer, including 58 lung, 60 pancreatic, and 59 gastrointestinal (GI) cancers, and 42 cancer‐free controls who died of other, non‐cardiovascular reasons. Cancer cachexia (CC) was defined based on clinical and/or pathological diagnosis, body mass index (BMI) <20.0 kg/m2 and/or oedema‐free body weight loss of 5.0% during the previous year or less. The pathology reports were analysed for BMI, heart weight (HW), and left and right ventricular wall thicknesses (LVWT and RVWT, respectively). The analysis of clinical data included recording of biochemical parameters and medication data of study patients. CC was detected in 54 (30.5%) subjects. Individuals with CC had a significantly lower HW than non‐cachectic subjects (363.1 ± 86.2 vs. 447.0 ± 128.9 g, P < 0.001) and control group (412.9 ± 75.8 g, P < 0.05). BMI correlated with HW in cases with GI cancer (r = 0.44, P < 0.001), lung cancer (r = 0.53, P < 0.0001), and pancreatic cancer (r = 0.39, P < 0.01). Conclusions Body weight loss in individuals with lung, pancreatic, and GI cancers is accompanied by a decrease in HW. In patients with CC who receive cancer treatment, screening for cardiac muscle wasting may have clinical importance.
Collapse
Affiliation(s)
- Anush Barkhudaryan
- Clinic of General and Invasive Cardiology, University Clinical Hospital No. 1, Yerevan, Armenia
| | - Nadja Scherbakov
- Center for Stroke Research Berlin, Charite Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Jochen Springer
- Institute of Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Centre Göttingen (UMG), Göttingen, Germany
| | - Wolfram Doehner
- Center for Stroke Research Berlin, Charite Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany.,Department of Cardiology and Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow, Charite Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
79
|
Thackeray JT, Pietzsch S, Stapel B, Ricke-Hoch M, Lee CW, Bankstahl JP, Scherr M, Heineke J, Scharf G, Haghikia A, Bengel FM, Hilfiker-Kleiner D. Insulin supplementation attenuates cancer-induced cardiomyopathy and slows tumor disease progression. JCI Insight 2017; 2:93098. [PMID: 28515362 DOI: 10.1172/jci.insight.93098] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/11/2017] [Indexed: 12/18/2022] Open
Abstract
Advanced cancer induces fundamental changes in metabolism and promotes cardiac atrophy and heart failure. We discovered systemic insulin deficiency in cachectic cancer patients. Similarly, mice with advanced B16F10 melanoma (B16F10-TM) or colon 26 carcinoma (C26-TM) displayed decreased systemic insulin associated with marked cardiac atrophy, metabolic impairment, and function. B16F10 and C26 tumors decrease systemic insulin via high glucose consumption, lowering pancreatic insulin production and producing insulin-degrading enzyme. As tumor cells consume glucose in an insulin-independent manner, they shift glucose away from cardiomyocytes. Since cardiomyocytes in both tumor models remained insulin responsive, low-dose insulin supplementation by subcutaneous implantation of insulin-releasing pellets improved cardiac glucose uptake, atrophy, and function, with no adverse side effects. In addition, by redirecting glucose to the heart in addition to other organs, the systemic insulin treatment lowered glucose usage by the tumor and thereby decreased tumor growth and volume. Insulin corrected the cancer-induced reduction in cardiac Akt activation and the subsequent overactivation of the proteasome and autophagy. Thus, cancer-induced systemic insulin depletion contributes to cardiac wasting and failure and may promote tumor growth. Low-dose insulin supplementation attenuates these processes and may be supportive in cardio-oncologic treatment concepts.
Collapse
Affiliation(s)
| | - Stefan Pietzsch
- Division of Molecular Cardiology, Department of Cardiology and Angiology, and
| | - Britta Stapel
- Division of Molecular Cardiology, Department of Cardiology and Angiology, and
| | - Melanie Ricke-Hoch
- Division of Molecular Cardiology, Department of Cardiology and Angiology, and
| | - Chun-Wei Lee
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | - Michaela Scherr
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Jörg Heineke
- Division of Molecular Cardiology, Department of Cardiology and Angiology, and
| | - Gesine Scharf
- Division of Molecular Cardiology, Department of Cardiology and Angiology, and
| | - Arash Haghikia
- Division of Molecular Cardiology, Department of Cardiology and Angiology, and.,Department of Cardiology, Charité Universitätsmedizin Berlin (Campus Benjamin Franklin), Berlin, Germany
| | | | | |
Collapse
|
80
|
Belloum Y, Rannou-Bekono F, Favier FB. Cancer-induced cardiac cachexia: Pathogenesis and impact of physical activity (Review). Oncol Rep 2017; 37:2543-2552. [PMID: 28393216 DOI: 10.3892/or.2017.5542] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/30/2017] [Indexed: 11/06/2022] Open
Abstract
Cachexia is a wasting syndrome observed in many patients suffering from several chronic diseases including cancer. In addition to the progressive loss of skeletal muscle mass, cancer cachexia results in cardiac function impairment. During the severe stage of the disease, patients as well as animals bearing cancer cells display cardiac atrophy. Cardiac energy metabolism is also impeded with disruption of mitochondrial homeostasis and reduced oxidative capacity, although the available data remain equivocal. The release of inflammatory cytokines by tumor is a key mechanism in the initiation of heart failure. Oxidative stress, which results from the combination of chemotherapy, inadequate antioxidant consumption and chronic inflammation, will further foster heart failure. Protein catabolism is due to the concomitant activation of proteolytic systems and inhibition of protein synthesis, both processes being triggered by the deactivation of the Akt/mammalian target of rapamycin pathway. The reduction in oxidative capacity involves AMP-activated protein kinase and peroxisome proliferator-activated receptor gamma coactivator 1α dysregulation. The nuclear factor-κB transcription factor plays a prominent role in the coordination of these alterations. Physical exercise appears as an interesting non-pharmaceutical way to counteract cancer cachexia-induced-heart failure. Indeed, aerobic training has anti-inflammatory effects, increases anti-oxidant defenses, prevents atrophy and promotes oxidative metabolism. The present review points out the importance of better understanding the concurrent structural and metabolic changes within the myocardium during cancer and the protective effects of exercise against cardiac cachexia.
Collapse
Affiliation(s)
| | - Françoise Rannou-Bekono
- EA 1274, Laboratoire 'Mouvement, Sport, Santé', Université de Rennes 2-ENS Rennes, Bruz 35170, France
| | | |
Collapse
|
81
|
Abstract
Functional changes in the heart in patients with cancer can be a result of both the disease itself and various cancer therapies, and limiting cardiac damage has become an increasingly important issue as survival rates in patients with cancer have improved. Processes involved in cancer-induced cardiac atrophy may include cardiomyocyte atrophy and apoptosis, decreased protein synthesis, increased autophagy and proteolysis via the ubiquitin-proteosome system. Further to direct effects of malignancy on the heart, several chemotherapeutic agents are known to affect the myocardium, in particular the anthracyclines. The aim of this report is to review the effects of cancer and cancer treatment on the heart and what is known about the underlying mechanisms. Furthermore, clinical strategies to limit and treat cancer-associated cardiac atrophy are discussed, emphasising the benefit of a multidisciplinary approach by cardiologists and oncologists to optimise models of care to improve outcomes for patients with cancer.
Collapse
Affiliation(s)
- Mark Sweeney
- Cardio-Oncology Service, Royal Brompton Hospital, London, UK
| | - Angela Yiu
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, UK
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, London, UK.,Faculty of Medicine, National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|
82
|
VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc Natl Acad Sci U S A 2016; 113:13144-13149. [PMID: 27799559 DOI: 10.1073/pnas.1616168113] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Congestive heart failure is one of the leading causes of disability in long-term survivors of cancer. The anthracycline antibiotic doxorubicin (DOX) is used to treat a variety of cancers, but its utility is limited by its cumulative cardiotoxicity. As advances in cancer treatment have decreased cancer mortality, DOX-induced cardiomyopathy has become an increasing problem. However, the current means to alleviate the cardiotoxicity of DOX are limited. We considered that vascular endothelial growth factor-B (VEGF-B), which promotes coronary arteriogenesis, physiological cardiac hypertrophy, and ischemia resistance, could be an interesting candidate for prevention of DOX-induced cardiotoxicity and congestive heart failure. To study this, we administered an adeno-associated viral vector expressing VEGF-B or control vector to normal and tumor-bearing mice 1 wk before DOX treatment, using doses mimicking the concentrations used in the clinics. VEGF-B treatment completely inhibited the DOX-induced cardiac atrophy and whole-body wasting. VEGF-B also prevented capillary rarefaction in the heart and improved endothelial function in DOX-treated mice. VEGF-B also protected cultured endothelial cells from apoptosis and restored their tube formation. VEGF-B increased left ventricular volume without compromising cardiac function, reduced the expression of genes associated with pathological remodeling, and improved cardiac mitochondrial respiration. Importantly, VEGF-B did not affect serum or tissue concentrations of DOX or augment tumor growth. By inhibiting DOX-induced endothelial damage, VEGF-B could provide a novel therapeutic possibility for the prevention of chemotherapy-associated cardiotoxicity in cancer patients.
Collapse
|
83
|
Boerma M, Sridharan V, Mao XW, Nelson GA, Cheema AK, Koturbash I, Singh SP, Tackett AJ, Hauer-Jensen M. Effects of ionizing radiation on the heart. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 770:319-327. [PMID: 27919338 DOI: 10.1016/j.mrrev.2016.07.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 12/20/2022]
Abstract
This article provides an overview of studies addressing effects of ionizing radiation on the heart. Clinical studies have identified early and late manifestations of radiation-induced heart disease, a side effect of radiation therapy to tumors in the chest when all or part of the heart is situated in the radiation field. Studies in preclinical animal models have contributed to our understanding of the mechanisms by which radiation may injure the heart. More recent observations in human subjects suggest that ionizing radiation may have cardiovascular effects at lower doses than was previously thought. This has led to examinations of low-dose photons and low-dose charged particle irradiation in animal models. Lastly, studies have started to identify non-invasive methods for detection of cardiac radiation injury and interventions that may prevent or mitigate these adverse effects. Altogether, this ongoing research should increase our knowledge of biological mechanisms of cardiovascular radiation injury, identify non-invasive biomarkers for early detection, and potential interventions that may prevent or mitigate these adverse effects.
Collapse
Affiliation(s)
- Marjan Boerma
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, AR, United States.
| | - Vijayalakshmi Sridharan
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, AR, United States
| | - Xiao-Wen Mao
- Loma Linda University, Department of Basic Sciences, Loma Linda, CA, United States
| | - Gregory A Nelson
- Loma Linda University, Department of Basic Sciences, Loma Linda, CA, United States
| | - Amrita K Cheema
- Georgetown University Medical Center, Departments of Oncology and Biochemistry, Molecular and Cellular Biology, Washington, DC, United States
| | - Igor Koturbash
- University of Arkansas for Medical Sciences, Department of Environment and Occupational Health, Little Rock, AR, United States
| | - Sharda P Singh
- University of Arkansas for Medical Sciences, Department of Pharmacology and Toxicology, Little Rock, AR, United States
| | - Alan J Tackett
- University of Arkansas for Medical Sciences, Department of Biochemistry and Molecular Biology, Little Rock, AR, United States
| | - Martin Hauer-Jensen
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, AR, United States; Central Arkansas Veterans Healthcare System, Surgical Service, Little Rock, AR, United States
| |
Collapse
|