1
|
Guo X, Zuo Z, Wang X, Sun Y, Xu D, Liu G, Tong Y, Zhang Z. Epidemiology, risk factors and mechanism of breast cancer and atrial fibrillation. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:92. [PMID: 39716319 DOI: 10.1186/s40959-024-00298-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Cancer and cardiovascular diseases are leading causes of death worldwide. Among them, breast cancer is one of the most common malignancies in women, while atrial fibrillation is one of the most extensively studied arrhythmias, with significant public health implications. As the global population ages and advancements in cancer treatments continue, the survival rates of breast cancer patients have significantly improved, leading to an increasing coexistence of breast cancer and atrial fibrillation. However, the mechanisms underlying this coexistence remain insufficiently studied, and there is no consensus on the optimal treatment strategies for these patients. This review consolidates existing research to systematically explore the epidemiological characteristics, risk factors, and pathophysiological mechanisms of both breast cancer and atrial fibrillation. It focuses on the unique signaling pathways associated with different molecular subtypes of breast cancer and their potential impact on the mechanisms of atrial fibrillation. Additionally, the relationship between atrial fibrillation treatment medications and breast cancer is discussed. These insights not only provide essential evidence for the precise prevention and management of atrial fibrillation in breast cancer patients but also lay a solid theoretical foundation for interdisciplinary clinical management practices.
Collapse
Affiliation(s)
- Xiaoxue Guo
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zheng Zuo
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Xishu Wang
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Ying Sun
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Dongyang Xu
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Guanghui Liu
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yi Tong
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zhiguo Zhang
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
2
|
Slavcheva SE, Angelov A. HER2-Targeted Therapy-From Pathophysiology to Clinical Manifestation: A Narrative Review. J Cardiovasc Dev Dis 2023; 10:489. [PMID: 38132657 PMCID: PMC10743885 DOI: 10.3390/jcdd10120489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Trastuzumab is the primary treatment for all stages of HER2-overexpressing breast cancer in patients. Though discovered over 20 years ago, trastuzumab-induced cardiotoxicity (TIC) remains a research topic in cardio-oncology. This review explores the pathophysiological basis of TIC and its clinical manifestations. Their understanding is paramount for early detection and cardioprotective treatment. Trastuzumab renders cardiomyocytes susceptible by inhibiting the cardioprotective NRG-1/HER2/HER4 signaling pathway. The drug acts on HER2-receptor-expressing cardiomyocytes, endothelium, and cardiac progenitor cells (see the Graphical Abstract). The activation of immune cells, fibroblasts, inflammation, and neurohormonal systems all contribute to the evolution of TIC. A substantial amount of research demonstrates that trastuzumab induces overt and subclinical left ventricular (LV) systolic failure. Data suggest the development of right ventricular damage, LV diastolic dysfunction, and heart failure with preserved ejection fraction. Further research is needed to define a chronological sequence of cardiac impairments to guide the proper timing of cardioprotection implementation.
Collapse
Affiliation(s)
- Svetoslava Elefterova Slavcheva
- First Department of Internal Diseases, EC Cardiology, Faculty of Medicine, Medical University “Prof. Dr. Paraskev Stoyanov”, 9000 Varna, Bulgaria;
- First Cardiology Clinic with Intensive Cardiology Activity, University Multiprofessional Hospital of Active Treatment “St. Marina”, 9000 Varna, Bulgaria
| | - Atanas Angelov
- First Department of Internal Diseases, EC Cardiology, Faculty of Medicine, Medical University “Prof. Dr. Paraskev Stoyanov”, 9000 Varna, Bulgaria;
- First Cardiology Clinic with Intensive Cardiology Activity, University Multiprofessional Hospital of Active Treatment “St. Marina”, 9000 Varna, Bulgaria
| |
Collapse
|
3
|
Kinoshita T, Onda N, Ohno R, Ikeda T, Sugizaki Y, Ohara H, Nakagami T, Yuzawa H, Shimada H, Shimizu K, Ikeda T. Activation recovery interval as an electrocardiographic repolarization index to detect doxorubicin-induced cardiotoxicity. J Cardiol 2023; 82:473-480. [PMID: 37506822 DOI: 10.1016/j.jjcc.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/12/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND It has been reported that early detection and treatment of cancer therapy- related cardiac dysfunction (CTRCD) improves its prognosis. The detailed relationships between electrocardiographic repolarization indices and decreased left ventricular function in CTRCD have not been elucidated. We closely assessed such relationships in patients with doxorubicin (DOX)-induced CTRCD. METHODS This retrospective, single-center, cohort study included 471 consecutive patients with malignant lymphoma who received chemotherapy including DOX. Of them, 17 patients with CTRCD and 68 patients without CTRCD who underwent 12‑lead electrocardiogram and an echocardiogram before and after chemotherapy were eventually analyzed. The fluctuations of the following electrocardiographic repolarization indices were evaluated in lead V5: QT, JT, T peak to T end interval (Tp-e), and activation recovery interval (ARI). These indices were corrected by heart rate with the Fridericia formula. RESULTS The median period from the end of chemotherapy to the diagnosis of the CTRCD group was 346 days (IQR 170-1283 days). After chemotherapy, the QT interval was significantly prolonged in both with and without CTRCD groups compared with that before chemotherapy (pre QTc vs. post QTc in CTRCD group, 386 ± 27 ms vs. 411 ± 37 ms, p = 0.03, pre QTc vs. post QTc in non-CTRCD group, 388 ± 24 ms vs. 395 ± 25 ms, p = 0.04, respectively). ARIc after chemotherapy was characteristically observed only in the CTRCD group (pre ARIc vs. post ARIc in CTRCD group, 258 ± 53 ms vs. 211 ± 28 ms, p = 0.03, pre ARIc vs. post ARIc in non-CTRCD group, 221 ± 19 ms vs. 225 ± 23 ms, NS, respectively) and had negative correlations with left ventricular ejection fraction (r = -0.56, p < 0.001). Using the receiver-operating characteristic curve, the relationship between ARIc and CTRCD morbidity was examined. The optimal cut-off point of ARIc prolongation between before and after chemotherapy was 18 ms (sensitivity 75 %, specificity 79 %, area under the curve 0.76). CONCLUSIONS ARIc prolongation may be useful in the early detection of developing late-onset chronic DOX-induced CTRCD and lead to early treatment for cardiac protection.
Collapse
Affiliation(s)
- Toshio Kinoshita
- Division of Cardiovascular Medicine, Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan.
| | - Naoki Onda
- Division of Hematology and Oncology, Department of Medicine, Toho University Omori Medical Center, Tokyo, Japan
| | - Ruiko Ohno
- Division of Cardiovascular Medicine, Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan
| | - Takushi Ikeda
- Division of Cardiovascular Medicine, Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan
| | - Yuta Sugizaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan
| | - Hiroshi Ohara
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, Tokyo, Japan
| | - Takahiro Nakagami
- Division of Cardiovascular Medicine, Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan
| | - Hitomi Yuzawa
- Division of Cardiology, Mitsui Memorial Hospital, Tokyo, Japan
| | - Hideaki Shimada
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhiro Shimizu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan
| | - Takanori Ikeda
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Wang TH, Ma Y, Gao S, Zhang WW, Han D, Cao F. Recent Advances in the Mechanisms of Cell Death and Dysfunction in Doxorubicin Cardiotoxicity. Rev Cardiovasc Med 2023; 24:336. [PMID: 39076437 PMCID: PMC11272847 DOI: 10.31083/j.rcm2411336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/26/2023] [Accepted: 06/12/2023] [Indexed: 07/31/2024] Open
Abstract
Despite recent advances in cancer therapy, anthracycline-based combination therapy remains the standardized first-line strategy and has been found to have effective antitumor actions. Anthracyclines are extremely cardiotoxic, which limits the use of these powerful chemotherapeutic agents. Although numerous studies have been conducted on the cardiotoxicity of anthracyclines, the precise mechanisms by which doxorubicin causes cardiomyocyte death and myocardial dysfunction remain incompletely understood. This review highlights recent updates in mechanisms and therapies involved in doxorubicin-induced cardiomyocyte death, including autophagy, ferroptosis, necroptosis, pyroptosis, and apoptosis, as well as mechanisms of cardiovascular dysfunction resulting in myocardial atrophy, defects in calcium handling, thrombosis, and cell senescence. We sought to uncover potential therapeutic approaches to manage anthracycline cardiotoxicity via manipulation of crucial targets involved in doxorubicin-induced cardiomyocyte death and dysfunction.
Collapse
Affiliation(s)
- Tian-Hu Wang
- National Clinical Research Center for Geriatric Diseases, the Second Medical Center, Chinese PLA
General Hospital, 100853 Beijing, China
| | - Yan Ma
- National Clinical Research Center for Geriatric Diseases, the Second Medical Center, Chinese PLA
General Hospital, 100853 Beijing, China
| | - Shan Gao
- National Clinical Research Center for Geriatric Diseases, the Second Medical Center, Chinese PLA
General Hospital, 100853 Beijing, China
| | - Wei-Wei Zhang
- National Clinical Research Center for Geriatric Diseases, the Second Medical Center, Chinese PLA
General Hospital, 100853 Beijing, China
| | - Dong Han
- National Clinical Research Center for Geriatric Diseases, the Second Medical Center, Chinese PLA
General Hospital, 100853 Beijing, China
| | - Feng Cao
- National Clinical Research Center for Geriatric Diseases, the Second Medical Center, Chinese PLA
General Hospital, 100853 Beijing, China
| |
Collapse
|
5
|
Chitrangi S, Vaity P, Jamdar A, Bhatt S. Patient-derived organoids for precision oncology: a platform to facilitate clinical decision making. BMC Cancer 2023; 23:689. [PMID: 37479967 PMCID: PMC10362580 DOI: 10.1186/s12885-023-11078-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/16/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Despite recent advances in research, there are still critical lacunae in our basic understanding of the cause, pathogenesis, and natural history of many cancers, especially heterogeneity in patient response to drugs and mediators in the transition from malignant to invasive phenotypes. The explication of the pathogenesis of cancer has been constrained by limited access to patient samples, tumor heterogeneity and lack of reliable biological models. Amelioration in cancer treatment depends on further understanding of the etiologic, genetic, biological, and clinical heterogeneity of tumor microenvironment. Patient-derived organoids recapitulate the basic features of primary tumors, including histological complexity and genetic heterogeneity, which is instrumental in predicting patient response to drugs. METHODS Human iPSCs from healthy donors, breast and ovarian cancer patients were successfully differentiated towards isogenic hepatic, cardiac, neural and endothelial lineages. Multicellular organoids were established using Primary cells isolated from tumor tissues, histologically normal tissues adjacent to the tumors (NATs) and adipose tissues (source of Mesenchymal Stem Cells) from ovarian and breast cancer patients. Further these organoids were propagated and used for drug resistance/sensitivity studies. RESULTS Ovarian and breast cancer patients' organoids showed heterogeneity in drug resistance and sensitivity. iPSCs-derived cardiomyocytes, hepatocytes and neurons showed donor-to-donor variability of chemotherapeutic drug sensitivity in ovarian cancer patients, breast cancer patients and healthy donors. CONCLUSION We report development of a novel integrated platform to facilitate clinical decision-making using the patient's primary cells, iPSCs and derivatives, to clinically relevant models for oncology research.
Collapse
Affiliation(s)
- Swati Chitrangi
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India
| | - Pooja Vaity
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India
| | - Aishwarya Jamdar
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India
| | - Shweta Bhatt
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India.
- Yashraj Biotechnology GmbH, Uhlandstraße 20-25, 10623, Berlin, Germany.
- Yashraj Biotechnology Limited, 8, The Green STE A, Dover, Delaware State, 19901, USA.
| |
Collapse
|
6
|
Desgres M, Lima Correa B, Petrusca L, Autret G, Pezzana C, Marigny C, Guillas C, Bellamy V, Vilar J, Perier MC, Dingli F, Loew D, Humbert C, Larghero J, Churlaud G, Renault N, Croisille P, Hagège A, Silvestre JS, Menasché P. Therapeutic potential of extracellular vesicles derived from cardiac progenitor cells in rodent models of chemotherapy-induced cardiomyopathy. Front Cardiovasc Med 2023; 10:1206279. [PMID: 37485274 PMCID: PMC10360184 DOI: 10.3389/fcvm.2023.1206279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
Background Current treatments of chemotherapy-induced cardiomyopathy (CCM) are of limited efficacy. We assessed whether repeated intravenous injections of human extracellular vesicles from cardiac progenitor cells (EV-CPC) could represent a new therapeutic option and whether EV manufacturing according to a Good Manufacturing Practices (GMP)-compatible process did not impair their bioactivity. Methods Immuno-competent mice received intra-peritoneal injections (IP) of doxorubicin (DOX) (4 mg/kg each; cumulative dose: 12 mg/kg) and were then intravenously (IV) injected three times with EV-CPC (total dose: 30 billion). Cardiac function was assessed 9-11 weeks later by cardiac magnetic resonance imaging (CMR) using strain as the primary end point. Then, immuno-competent rats received 5 IP injections of DOX (3 mg/kg each; cumulative dose 15 mg/kg) followed by 3 equal IV injections of GMP-EV (total dose: 100 billion). Cardiac function was assessed by two dimensional-echocardiography. Results In the chronic mouse model of CCM, DOX + placebo-injected hearts incurred a significant decline in basal (global, epi- and endocardial) circumferential strain compared with sham DOX-untreated mice (p = 0.043, p = 0.042, p = 0.048 respectively) while EV-CPC preserved these indices. Global longitudinal strain followed a similar pattern. In the rat model, IV injections of GMP-EV also preserved left ventricular end-systolic and end-diastolic volumes compared with untreated controls. Conclusions Intravenously-injected extracellular vesicles derived from CPC have cardio-protective effects which may make them an attractive user-friendly option for the treatment of CCM.
Collapse
Affiliation(s)
| | | | - Lorena Petrusca
- Université de Lyon, INSA, Université Claude Bernard Lyon 1, UJM-Saint-Etienne, CNRS UMR 5520, INSERM U1206, CREATIS, Saint-Etienne, France
| | - Gwennhael Autret
- Université Paris Cité, Inserm, PARCC, Paris, France
- Plateforme Imageries du Vivant, Université Paris Cité, UFR de médecine, Paris, France
| | | | | | | | | | - José Vilar
- Université Paris Cité, Inserm, PARCC, Paris, France
| | | | - Florent Dingli
- Institut Curie, PSL Research University, Centre de Recherche, Curie CoreTech Mass Spectrometry Proteomics, Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Curie CoreTech Mass Spectrometry Proteomics, Paris, France
| | - Camille Humbert
- MEARY Cell and Gene Therapy Center, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jérôme Larghero
- Université Paris Cité, AP-HP, Hôpital Saint-Louis, MEARY Cell and Gene Therapy Center, Hôpital Saint Louis, INSERM CIC-BT CBT501, Paris, France
| | - Guillaume Churlaud
- MEARY Cell and Gene Therapy Center, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Nisa Renault
- FUJIFILM Cellular Dynamics, Inc., Madison, WI, United States
| | - Pierre Croisille
- Université de Lyon, INSA, Université Claude Bernard Lyon 1, UJM-Saint-Etienne, CNRS UMR 5520, INSERM U1206, CREATIS, Saint-Etienne, France
| | - Albert Hagège
- Université Paris Cité, Inserm, PARCC, Paris, France
- Department of Cardiology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | | | - Philippe Menasché
- Université Paris Cité, Inserm, PARCC, Paris, France
- Department of Cardiovascular Surgery, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
7
|
Leiva O, Bohart I, Ahuja T, Park D. Off-Target Effects of Cancer Therapy on Development of Therapy-Induced Arrhythmia: A Review. Cardiology 2023; 148:324-334. [PMID: 36702116 PMCID: PMC10614257 DOI: 10.1159/000529260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Advances in cancer therapeutics have improved overall survival and prognosis in this patient population; however, this has come at the expense of cardiotoxicity including arrhythmia. SUMMARY Cancer and its therapies are associated with cardiotoxicity via several mechanisms including inflammation, cardiomyopathy, and off-target effects. Among cancer therapies, anthracyclines and tyrosine kinase inhibitors (TKIs) are particularly known for their pro-arrhythmia effects. In addition to cardiomyopathy, anthracyclines may be pro-arrhythmogenic via reactive oxygen species (ROS) generation and altered calcium handling. TKIs may mediate their cardiotoxicity via inhibition of off-target tyrosine kinases. Ibrutinib-mediated inhibition of CSK may be responsible for the increased prevalence of atrial fibrillation. Further investigation is warranted to further elucidate the mechanisms behind arrhythmias in cancer therapies. KEY MESSAGES Arrhythmias are a common cardiotoxicity of cancer therapies. Cancer therapies may induce arrhythmias via off-target effects. Understanding the mechanisms underlying arrhythmogenesis associated with cancer therapies may help design cancer therapies that can avoid these toxicities.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Isaac Bohart
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Tania Ahuja
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - David Park
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
8
|
Moro N, Dokshokova L, Perumal Vanaja I, Prando V, Cnudde SJA, Di Bona A, Bariani R, Schirone L, Bauce B, Angelini A, Sciarretta S, Ghigo A, Mongillo M, Zaglia T. Neurotoxic Effect of Doxorubicin Treatment on Cardiac Sympathetic Neurons. Int J Mol Sci 2022; 23:ijms231911098. [PMID: 36232393 PMCID: PMC9569551 DOI: 10.3390/ijms231911098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 11/26/2022] Open
Abstract
Doxorubicin (DOXO) remains amongst the most commonly used anti-cancer agents for the treatment of solid tumors, lymphomas, and leukemias. However, its clinical use is hampered by cardiotoxicity, characterized by heart failure and arrhythmias, which may require chemotherapy interruption, with devastating consequences on patient survival and quality of life. Although the adverse cardiac effects of DOXO are consolidated, the underlying mechanisms are still incompletely understood. It was previously shown that DOXO leads to proteotoxic cardiomyocyte (CM) death and myocardial fibrosis, both mechanisms leading to mechanical and electrical dysfunction. While several works focused on CMs as the culprits of DOXO-induced arrhythmias and heart failure, recent studies suggest that DOXO may also affect cardiac sympathetic neurons (cSNs), which would thus represent additional cells targeted in DOXO-cardiotoxicity. Confocal immunofluorescence and morphometric analyses revealed alterations in SN innervation density and topology in hearts from DOXO-treated mice, which was consistent with the reduced cardiotropic effect of adrenergic neurons in vivo. Ex vivo analyses suggested that DOXO-induced denervation may be linked to reduced neurotrophic input, which we have shown to rely on nerve growth factor, released from innervated CMs. Notably, similar alterations were observed in explanted hearts from DOXO-treated patients. Our data demonstrate that chemotherapy cardiotoxicity includes alterations in cardiac innervation, unveiling a previously unrecognized effect of DOXO on cardiac autonomic regulation, which is involved in both cardiac physiology and pathology, including heart failure and arrhythmias.
Collapse
Affiliation(s)
- Nicola Moro
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Lolita Dokshokova
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Induja Perumal Vanaja
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Valentina Prando
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Sophie Julie A Cnudde
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Riccardo Bariani
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza, University of Rome, 04100 Latina, Italy
| | - Barbara Bauce
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Annalisa Angelini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza, University of Rome, 04100 Latina, Italy
| | - Alessandra Ghigo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.M.); (T.Z.); Tel.: +39-0497923229 (M.M.); +39-0497923294 (T.Z.); Fax: +39-0497923250 (M.M.); +39-0497923250 (T.Z.)
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.M.); (T.Z.); Tel.: +39-0497923229 (M.M.); +39-0497923294 (T.Z.); Fax: +39-0497923250 (M.M.); +39-0497923250 (T.Z.)
| |
Collapse
|
9
|
Yu L, Jin Y, Song M, Zhao Y, Zhang H. When Natural Compounds Meet Nanotechnology: Nature-Inspired Nanomedicines for Cancer Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14081589. [PMID: 36015215 PMCID: PMC9412684 DOI: 10.3390/pharmaceutics14081589] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Recent significant strides of natural compounds in immunomodulation have highlighted their great potential against cancer. Despite many attempts being made for cancer immunotherapy, the biomedical application of natural compounds encounters a bottleneck because of their unclear mechanisms, low solubility and bioavailability, and limited efficacy. Herein, we summarize the immune regulatory mechanisms of different natural compounds at each step of the cancer-immunity cycle and highlight their anti-tumor potential and current limitations. We then propose and present various drug delivery strategies based on nanotechnology, including traditional nanoparticles (NPs)-based delivery strategies (lipid-based NPs, micelles, and polysaccharide/peptide/protein-based NPs) and novel delivery strategies (cell-derived NPs and carrier-free NPs), thus providing solutions to break through existing bottlenecks. Furthermore, representative applications of nature-inspired nanomedicines are also emphasized in detail with the advantages and disadvantages discussed. Finally, the challenges and prospects of natural compounds for cancer immunotherapy are provided, hopefully, to facilitate their far-reaching development toward clinical translation.
Collapse
Affiliation(s)
- Linna Yu
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
| | - Yi Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Mingjie Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Yu Zhao
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
- Correspondence: (Y.Z.); (H.Z.)
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
- Correspondence: (Y.Z.); (H.Z.)
| |
Collapse
|
10
|
Meng C, Fan L, Wang X, Wang Y, Li Y, Pang S, Lv S, Zhang J. Preparation and Evaluation of Animal Models of Cardiotoxicity in Antineoplastic Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3820591. [PMID: 35847594 PMCID: PMC9277159 DOI: 10.1155/2022/3820591] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
Abstract
The continuous development of antineoplastic therapy has significantly reduced the mortality of patients with malignant tumors, but its induced cardiotoxicity has become the primary cause of long-term death in patients with malignant tumors. However, the pathogenesis of cardiotoxicity of antineoplastic therapy is currently unknown, and practical means of prevention and treatment are lacking in clinical practice. Therefore, how to effectively prevent and treat cardiotoxicity while treating tumors is a major challenge. Animal models are important tools for studying cardiotoxicity in antitumor therapy and are of great importance in elucidating pathophysiological mechanisms and developing and evaluating modality drugs. In this paper, we summarize the existing animal models in antitumor therapeutic cardiotoxicity studies and evaluate the models by observing the macroscopic signs, echocardiography, and pathological morphology of the animals, aiming to provide a reference for subsequent experimental development and clinical application.
Collapse
Affiliation(s)
- Chenchen Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Lu Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Xiaoming Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Yanyang Li
- Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
- Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| |
Collapse
|
11
|
Lazzarini E, Lodrini AM, Arici M, Bolis S, Vagni S, Panella S, Rendon-Angel A, Saibene M, Metallo A, Torre T, Vassalli G, Ameri P, Altomare C, Rocchetti M, Barile L. Stress-induced premature senescence is associated with a prolonged QT interval and recapitulates features of cardiac aging. Theranostics 2022; 12:5237-5257. [PMID: 35836799 PMCID: PMC9274748 DOI: 10.7150/thno.70884] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/11/2022] [Indexed: 01/12/2023] Open
Abstract
Rationale: Aging in the heart is a gradual process, involving continuous changes in cardiovascular cells, including cardiomyocytes (CMs), namely cellular senescence. These changes finally lead to adverse organ remodeling and resulting in heart failure. This study exploits CMs from human induced pluripotent stem cells (iCMs) as a tool to model and characterize mechanisms involved in aging. Methods and Results: Human somatic cells were reprogrammed into human induced pluripotent stem cells and subsequently differentiated in iCMs. A senescent-like phenotype (SenCMs) was induced by short exposure (3 hours) to doxorubicin (Dox) at the sub-lethal concentration of 0.2 µM. Dox treatment induced expression of cyclin-dependent kinase inhibitors p21 and p16, and increased positivity to senescence-associated beta-galactosidase when compared to untreated iCMs. SenCMs showed increased oxidative stress, alteration in mitochondrial morphology and depolarized mitochondrial membrane potential, which resulted in decreased ATP production. Functionally, when compared to iCMs, SenCMs showed, prolonged multicellular QTc and single cell APD, with increased APD variability and delayed afterdepolarizations (DADs) incidence, two well-known arrhythmogenic indexes. These effects were largely ascribable to augmented late sodium current (INaL) and reduced delayed rectifier potassium current (Ikr). Moreover sarcoplasmic reticulum (SR) Ca2+ content was reduced because of downregulated SERCA2 and increased RyR2-mediated Ca2+ leak. Electrical and intracellular Ca2+ alterations were mostly justified by increased CaMKII activity in SenCMs. Finally, SenCMs phenotype was furtherly confirmed by analyzing physiological aging in CMs isolated from old mice in comparison to young ones. Conclusions: Overall, we showed that SenCMs recapitulate the phenotype of aged primary CMs in terms of senescence markers, electrical and Ca2+ handling properties and metabolic features. Thus, Dox-induced SenCMs can be considered a novel in vitro platform to study aging mechanisms and to envision cardiac specific anti-aging approach in humans.
Collapse
Affiliation(s)
- Edoardo Lazzarini
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Alessandra Maria Lodrini
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milano, Italy.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Martina Arici
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milano, Italy
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Sara Vagni
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milano, Italy
| | - Stefano Panella
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Azucena Rendon-Angel
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Melissa Saibene
- Department of Earth and Environmental Sciences, Università degli Studi di Milano-Bicocca, Milano, Italy
| | - Alessia Metallo
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milano, Italy
| | - Tiziano Torre
- Department of Cardiac Surgery Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giuseppe Vassalli
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico, Genova, Italy.,Department of Internal Medicine, University of Genova, Genova, Italy
| | - Claudia Altomare
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milano, Italy.,✉ Corresponding authors: Lucio Barile, PhD. Istituto Cardiocentro Ticino, Laboratories for Translational Research, EOC Via Chiesa 5, 6500 Bellinzona, Switzerland. +41 586667104 ; Marcella Rocchetti, PhD. University of Milano-Bicocca, Dept. of Biotechnology and Biosciences, P.za della Scienza 2, 20126 Milano, Italy. +39 0264483313
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.,Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.,✉ Corresponding authors: Lucio Barile, PhD. Istituto Cardiocentro Ticino, Laboratories for Translational Research, EOC Via Chiesa 5, 6500 Bellinzona, Switzerland. +41 586667104 ; Marcella Rocchetti, PhD. University of Milano-Bicocca, Dept. of Biotechnology and Biosciences, P.za della Scienza 2, 20126 Milano, Italy. +39 0264483313
| |
Collapse
|
12
|
Pecoraro M, Marzocco S, Franceschelli S, Popolo A. Trastuzumab and Doxorubicin Sequential Administration Increases Oxidative Stress and Phosphorylation of Connexin 43 on Ser368. Int J Mol Sci 2022; 23:ijms23126375. [PMID: 35742818 PMCID: PMC9224207 DOI: 10.3390/ijms23126375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 02/05/2023] Open
Abstract
Human epidermal growth factor receptor-2 (HER2) is overexpressed in up to 30% of breast cancer cases, causing a more aggressive tumour growth and poor prognosis. Trastuzumab, the humanized antibody targeted to HER2, increased the life expectancy of patients, but severe cardiotoxicity emerged as a long-term adverse effect. Clinical evidence highlights that Trastuzumab-induced cardiotoxicity drastically increases in association with Doxorubicin; however, the exact mechanisms involved remain incompletely understood. In order to analyse the molecular mechanisms involved and the possible adaptative responses to Trastuzumab and Doxorubicin treatment, in this study, H9c2 cardiomyoblasts were used. Results showed that Trastuzumab and Doxorubicin sequential administration in cardiomyoblast increased cytosolic and mitochondrial ROS production, intracellular calcium dysregulation, mitochondrial membrane depolarization, and the consequent apoptosis, induced by both Trastuzumab and Doxorubicin alone. Furthermore, in these conditions, we observed increased levels of Connexin43 phosphorylated on Ser368 (pCx43). Since phosphorylation on Ser368 decreases gap junction intracellular communication, thus reducing the spread of death signals to adjacent cells, we hypothesized that the increase in pCx43 could be an adaptative response implemented by cells to defend neighbouring cells by Trastuzumab and Doxorubicin sequential administration. However, the other side of the coin is the resulting conduction abnormalities.
Collapse
|