1
|
Sayour NV, Paál ÁM, Ameri P, Meijers WC, Minotti G, Andreadou I, Lombardo A, Camilli M, Drexel H, Grove EL, Dan GA, Ivanescu A, Semb AG, Savarese G, Dobrev D, Crea F, Kaski JC, de Boer RA, Ferdinandy P, Varga ZV. Heart failure pharmacotherapy and cancer: pathways and pre-clinical/clinical evidence. Eur Heart J 2024; 45:1224-1240. [PMID: 38441940 PMCID: PMC11023004 DOI: 10.1093/eurheartj/ehae105] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 04/08/2024] Open
Abstract
Heart failure (HF) patients have a significantly higher risk of new-onset cancer and cancer-associated mortality, compared to subjects free of HF. While both the prevention and treatment of new-onset HF in patients with cancer have been investigated extensively, less is known about the prevention and treatment of new-onset cancer in patients with HF, and whether and how guideline-directed medical therapy (GDMT) for HF should be modified when cancer is diagnosed in HF patients. The purpose of this review is to elaborate and discuss the effects of pillar HF pharmacotherapies, as well as digoxin and diuretics on cancer, and to identify areas for further research and novel therapeutic strategies. To this end, in this review, (i) proposed effects and mechanisms of action of guideline-directed HF drugs on cancer derived from pre-clinical data will be described, (ii) the evidence from both observational studies and randomized controlled trials on the effects of guideline-directed medical therapy on cancer incidence and cancer-related outcomes, as synthetized by meta-analyses will be reviewed, and (iii) considerations for future pre-clinical and clinical investigations will be provided.
Collapse
Affiliation(s)
- Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Ágnes M Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Italian IRCCS Cardiology Network, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Wouter C Meijers
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Giorgio Minotti
- University Campus Bio-Medico, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Ioanna Andreadou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation & Treatment (VIVIT), Carinagasse 47, A-6800 Feldkirch, Austria
| | - Erik Lerkevang Grove
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Gheorghe Andrei Dan
- Carol Davila University of Medicine and Pharmacy, Colentina University Hospital, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Andreea Ivanescu
- Carol Davila University of Medicine and Pharmacy, Colentina University Hospital, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Anne Grete Semb
- Division of Research and Innovation, REMEDY-Centre for Treatment of Rheumatic and Musculoskeletal Diseases, Diakonhjemmet Hospital, Oslo, Norway
| | - Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular and Neuro Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, QC, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Juan-Carlos Kaski
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Rudolf A de Boer
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- Pharmahungary Group, Szeged, Hungary
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
| |
Collapse
|
2
|
Lee D, Ham IH, Oh HJ, Lee DM, Yoon JH, Son SY, Kim TM, Kim JY, Han SU, Hur H. Tubulointerstitial nephritis antigen-like 1 from cancer-associated fibroblasts contribute to the progression of diffuse-type gastric cancers through the interaction with integrin β1. J Transl Med 2024; 22:154. [PMID: 38355577 PMCID: PMC10868052 DOI: 10.1186/s12967-024-04963-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Tumor cells of diffuse-type gastric cancer (DGC) are discohesive and infiltrate into the stroma as single cells or small subgroups, so the stroma significantly impacts DGC progression. Cancer-associated fibroblasts (CAFs) are major components of the tumor stroma. Here, we identified CAF-specific secreted molecules and investigated the mechanism underlying CAF-induced DGC progression. METHODS We conducted transcriptome analysis for paired normal fibroblast (NF)-CAF isolated from DGC patient tissues and proteomics for conditioned media (CM) of fibroblasts. The effects of fibroblasts on cancer cells were examined by transwell migration and soft agar assays, western blotting, and in vivo. We confirmed the effect of blocking tubulointerstitial nephritis antigen-like 1 (TINAGL1) in CAFs using siRNA or shRNA. We evaluated the expression of TINAGL1 protein in frozen tissues of DGC and paired normal stomach and mRNA in formalin-fixed, paraffin-embedded (FFPE) tissue using RNA in-situ hybridization (RNA-ISH). RESULTS CAFs more highly expressed TINAGL1 than NFs. The co-culture of CAFs increased migration and tumorigenesis of DGC. Moreover, CAFs enhanced the phosphorylation of focal adhesion kinase (FAK) and mesenchymal marker expression in DGC cells. In an animal study, DGC tumors co-injected with CAFs showed aggressive phenotypes, including lymph node metastasis. However, increased phosphorylation of FAK and migration were reduced by blocking TINAGL1 in CAFs. In the tissues of DGC patients, TINAGL1 was higher in cancer than paired normal tissues and detected with collagen type I alpha 1 chain (COL1A1) in the same spot. Furthermore, high TINAGL1 expression was significantly correlated with poor prognosis in several public databases and our patient cohort diagnosed with DGC. CONCLUSIONS These results indicate that TINAGL1 secreted by CAFs induces phosphorylation of FAK in DGC cells and promotes tumor progression. Thus, targeting TINAGL1 in CAFs can be a novel therapeutic strategy for DGC.
Collapse
Affiliation(s)
- Dagyeong Lee
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Cancer Biology Graduate Program, Ajou University School of Medicine Suwon, Suwon, Republic of Korea
- AI-Super Convergence KIURI Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| | - In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Inflamm-Aging Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hye Jeong Oh
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dong Min Lee
- Inflamm-Aging Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea Seoul, Seoul, Republic of Korea
| | - Sang-Yong Son
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Science, Graduate School, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, Republic of Korea
| | - Sang-Uk Han
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea.
- Cancer Biology Graduate Program, Ajou University School of Medicine Suwon, Suwon, Republic of Korea.
- Inflamm-Aging Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
3
|
Zhao Y, Jia H, Hua X, An T, Song J. Cardio-oncology: Shared Genetic, Metabolic, and Pharmacologic Mechanism. Curr Cardiol Rep 2023; 25:863-878. [PMID: 37493874 PMCID: PMC10403418 DOI: 10.1007/s11886-023-01906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2023] [Indexed: 07/27/2023]
Abstract
PURPOSE OF REVIEW The article aims to investigate the complex relationship between cancer and cardiovascular disease (CVD), with a focus on the effects of cancer treatment on cardiac health. RECENT FINDINGS Advances in cancer treatment have improved long-term survival rates, but CVD has emerged as a leading cause of morbidity and mortality in cancer patients. The interplay between cancer itself, treatment methods, homeostatic changes, and lifestyle modifications contributes to this comorbidity. Recent research in the field of cardio-oncology has revealed common genetic mutations, risk factors, and metabolic features associated with the co-occurrence of cancer and CVD. This article provides a comprehensive review of the latest research in cardio-oncology, including common genetic mutations, risk factors, and metabolic features, and explores the interactions between cancer treatment and CVD drugs, proposing novel approaches for the management of cancer and CVD.
Collapse
Affiliation(s)
- Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Tao An
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| |
Collapse
|
4
|
Wawszczyk J, Wolan R, Smolik S, Kapral M. In vitro and in silico study on the effect of carvedilol and sorafenib alone and in combination on the growth and inflammatory response of melanoma cells. Saudi Pharm J 2023; 31:1306-1316. [PMID: 37323921 PMCID: PMC10265481 DOI: 10.1016/j.jsps.2023.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Melanoma is an aggressive skin cancer. Increasing evidence has shown the role of β-adrenergic receptors in the pathogenesis of melanoma. Carvedilol is a widely used non-selective β-AR antagonist with potential anticancer activity. The purpose of the study was to estimate the influence of carvedilol and sorafenib alone and in combination on the growth and inflammatory response of C32 and A2058 melanoma cells. Furthermore, this study also aimed to predict the probable interaction of carvedilol and sorafenib when administered together. Predictive study of the interaction of carvedilol and sorafenib was performed using the ChemDIS-Mixture system. Carvedilol and sorafenib alone and in combination showed a growth inhibitory effect on cells. The greatest synergistic antiproliferative effect on both cell lines was observed at Car 5 μM combined with Sor 5 μM. Analysis in silico identified diseases, proteins, and metabolic pathways that can be affected by the interaction of carvedilol and sorafenib. The results obtained demonstrated that carvedilol and sorafenib modulated the secretion of IL-8 by IL-1β-stimulated by melanoma cell lines but the use of a combination of both drugs did not intensify the effect. In summary, the results presented indicate that the combination of carvedilol and sorafenib may have a promising anticancer effect on melanoma cells.
Collapse
|
5
|
The β-Blocker Carvedilol Prevents Benzo(a)pyrene-Induced Lung Toxicity, Inflammation and Carcinogenesis. Cancers (Basel) 2023; 15:cancers15030583. [PMID: 36765542 PMCID: PMC9913110 DOI: 10.3390/cancers15030583] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
The current study evaluated the effects of the β-blocker carvedilol on benzo(a)pyrene (B(a)P) and its active metabolite benzo(a)pyrene diol epoxide (BPDE)-induced lung toxicity, inflammation and carcinogenesis and explored the potential mechanisms. Carvedilol blocked the BPDE-induced malignant transformation of human bronchial epithelial cells BEAS-2B. In BEAS-2B cells, B(a)P strongly activated ELK-1, a transcription factor regulating serum response element (SRE) signaling, which was attenuated by carvedilol. Carvedilol also inhibited the B(a)P-induced AhR/xenobiotic responsive element (XRE) and mRNA expression of CYP1A1 and attenuated B(a)P-induced NF-κB activation. In a B(a)P-induced acute lung toxicity model in CD-1/IGS mice, pretreatment with carvedilol for 7 days before B(a)P exposure effectively inhibited the B(a)P-induced plasma levels of lactate dehydrogenase and malondialdehyde, inflammatory cell infiltration and histopathologic abnormalities in the lung, and upregulated the expression of GADD45α, caspase-3 and COX-2 in the lung. In a B(a)P-induced lung carcinogenesis model in A/J mice, carvedilol treatment for 20 weeks did not affect body weight but significantly attenuated tumor multiplicity and volume. These data reveal a previously unexplored role of carvedilol in preventing B(a)P-induced lung inflammation and carcinogenesis by inhibiting the cross-talk of the oncogenic transcription factors ELK-1, AhR and NF-κB.
Collapse
|
6
|
Sauerbruch T, Hennenberg M, Trebicka J, Schierwagen R. Beta-blockers in patients with liver cirrhosis: Pragmatism or perfection? Front Med (Lausanne) 2023; 9:1100966. [PMID: 36743678 PMCID: PMC9891090 DOI: 10.3389/fmed.2022.1100966] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
With increasing decompensation, hyperdynamic circulatory disturbance occurs in liver cirrhosis despite activation of vasoconstrictors. Here, the concept of a therapy with non-selective beta-blockers was established decades ago. They lower elevated portal pressure, protect against variceal hemorrhage, and may also have pleiotropic immunomodulatory effects. Recently, the beneficial effect of carvedilol, which blocks alpha and beta receptors, has been highlighted. Carvedilol leads to "biased-signaling" via recruitment of beta-arrestin. This effect and its consequences have not been sufficiently investigated in patients with liver cirrhosis. Also, a number of questions remain open regarding the expression of beta-receptors and its intracellular signaling and the respective consequences in the intra- and extrahepatic tissue compartments. Despite the undisputed role of non-selective beta-blockers in the treatment of liver cirrhosis, we still can improve the knowledge as to when and how beta-blockers should be used in which patients.
Collapse
Affiliation(s)
- Tilman Sauerbruch
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Martin Hennenberg
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jonel Trebicka
- Department of Internal Medicine B, University of Münster, Münster, Germany
- European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain
| | - Robert Schierwagen
- Department of Internal Medicine B, University of Münster, Münster, Germany
| |
Collapse
|
7
|
Topical carvedilol delivery prevents UV-induced skin cancer with negligible systemic absorption. Int J Pharm 2022; 611:121302. [PMID: 34793935 PMCID: PMC8692451 DOI: 10.1016/j.ijpharm.2021.121302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 01/07/2023]
Abstract
The β-blocker carvedilol prevents ultraviolet (UV)-induced skin cancer, but systemic drug administration may cause unwanted cadiovascular effects. To overcome this limitation, a topical delivery system based on transfersome (T-CAR) was characterized ex vivo and in vivo. T-CAR was visualized by Transmission Electron Microscopy as nanoparticles of spherical and unilamellar structure. T-CAR incorporated into carbopol gel and in suspension showed similar drug permeation and deposition profiles in Franz diffusion cells loaded with porcine ear skin. In mice exposed to a single dose UV, topical T-CAR gel (10 µM) significantly reduced UV-induced skin edema and cyclobutane pyrimidine dimer formation. In mice exposed to chronic UV radiation for 25 weeks, topical T-CAR gel (10 µM) significantly delayed the incidence of tumors, reduced tumor number and burden, and attenuated Ki-67 and COX-2 expression. The T-CAR gel was subsequently examined for skin deposition, systemic absorption and cardiovascular effects in mice. In mice treated with repeated doses of T-CAR gel (100 µM), the drug was undetectable in plasma, the heart rate was unaffected, but skin deposition was significantly higher than mice treated with oral carvedilol (32 mg/kg/day). These data indicate that the carbopol-based T-CAR gel holds great promise for skin cancer prevention with negligible systemic effects.
Collapse
|
8
|
Effect of a Low Dose of Carvedilol on Cyclophosphamide-Induced Urinary Toxicity in Rats—A Comparison with Mesna. Pharmaceuticals (Basel) 2021; 14:ph14121237. [PMID: 34959638 PMCID: PMC8708009 DOI: 10.3390/ph14121237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/21/2021] [Accepted: 11/25/2021] [Indexed: 01/28/2023] Open
Abstract
One of the major side effects of cyclophosphamide (CPX)—an alkylating anticancer drug that is still clinically used—is urotoxicity with hemorrhagic cystitis. The present study was designed to evaluate the ability of carvedilol to protect rats from cyclophosphamide-induced urotoxicity. Rats were injected intraperitoneally (i.p.) with CPX (200 mg/kg) and administered carvedilol (2 mg/kg) intragastrically a day before, at the day and a day after a single i.p. injection of CPX, with or without mesna (40, 80, and 80 mg/kg i.p. 20 min before, 4 h and 8 h after CPX administration, respectively). Pretreatment with carvedilol partly prevented the CPX-induced increase in urinary bladder and kidney index, and completely protects from CPX-evoked alterations in serum potassium and creatinine level, but did not prevent histological alterations in the urinary bladder and hematuria. However, carvedilol administration resulted in significant restoration of kidney glutathione (GSH) level and a decrease in kidney interleukin 1β (IL-1β) and plasma asymmetric dimethylarginine (ADMA) concentrations. Not only did mesna improve kidney function, but it also completely reversed histological abnormalities in bladders and prevented hematuria. In most cases, no significant interaction of carvedilol with mesna was observed, although the effect of both drugs together was better than mesna given alone regarding plasma ADMA level and kidney IL-1β concentration. In conclusion, carvedilol did not counteract the injury caused in the urinary bladders but restored kidney function, presumably via its antioxidant and anti-inflammatory properties.
Collapse
|
9
|
Photosensitizing Medications and Skin Cancer: A Comprehensive Review. Cancers (Basel) 2021; 13:cancers13102344. [PMID: 34066301 PMCID: PMC8152064 DOI: 10.3390/cancers13102344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
(1) The incidence of skin cancer is increasing in the United States (US) despite scientific advances in our understanding of skin cancer risk factors and treatments. In vitro and in vivo studies have provided evidence that suggests that certain photosensitizing medications (PSMs) increase skin cancer risk. This review summarizes current epidemiological evidence on the association between common PSMs and skin cancer. (2) A comprehensive literature search was conducted to identify meta-analyses, observational studies and clinical trials that report on skin cancer events in PSM users. The associated risks of keratinocyte carcinoma (squamous cell carcinoma and basal cell carcinoma) and melanoma are summarized, for each PSM. (3) There are extensive reports on antihypertensives and statins relative to other PSMs, with positive and null findings, respectively. Fewer studies have explored amiodarone, metformin, antimicrobials and vemurafenib. No studies report on the individual skin cancer risks in glyburide, naproxen, piroxicam, chlorpromazine, thioridazine and nalidixic acid users. (4) The research gaps in understanding the relationship between PSMs and skin cancer outlined in this review should be prioritized because the US population is aging. Thus the number of patients prescribed PSMs is likely to continue to rise.
Collapse
|
10
|
Liang S, Shamim MA, Shahid A, Chen M, Cleveland KH, Parsa C, Orlando R, Andresen BT, Huang Y. Prevention of Skin Carcinogenesis by the Non-β-blocking R-carvedilol Enantiomer. Cancer Prev Res (Phila) 2021; 14:527-540. [PMID: 33648941 DOI: 10.1158/1940-6207.capr-20-0609] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/22/2021] [Accepted: 02/25/2021] [Indexed: 11/16/2022]
Abstract
Skin cancer is the most common malignancy worldwide and is rapidly rising in incidence, representing a significant public health challenge. The β-blocker, carvedilol, has shown promising effects in preventing skin cancer. However, as a potent β-blocker, repurposing carvedilol to an anticancer agent is limited by cardiovascular effects. Carvedilol is a racemic mixture consisting of equimolar S- and R-carvedilol, whereas the R-carvedilol enantiomer does not possess β-blocking activity. Because previous studies suggest that carvedilol's cancer preventive activity is independent of β-blockade, we examined the skin cancer preventive activity of R-carvedilol compared with S-carvedilol and the racemic carvedilol. R- and S-carvedilol were equally effective in preventing EGF-induced neoplastic transformation of the mouse epidermal JB6 Cl 41-5a (JB6 P+) cells and displayed similar attenuation of EGF-induced ELK-1 activity. R-carvedilol appeared slightly better than S-carvedilol against UV-induced intracellular oxidative stress and release of prostaglandin E2 from the JB6 P+ cells. In an acute UV-induced skin damage and inflammation mouse model using a single irradiation of 300 mJ/cm2 UV, topical treatment with R-carvedilol dose dependently attenuated skin edema and reduced epidermal thickening, Ki-67 staining, COX-2 protein, and IL6 and IL1β mRNA levels similar to carvedilol. In a chronic UV (50-150 mJ/cm2) induced skin carcinogenesis model in mice with pretreatment of test agents, topical treatment with R-carvedilol, but not racemic carvedilol, significantly delayed and reduced skin squamous cell carcinoma development. Therefore, as an enantiomer present in an FDA-approved agent, R-carvedilol may be a better option for developing a safer and more effective preventive agent for skin carcinogenesis. PREVENTION RELEVANCE: In this study, we demonstrated the skin cancer preventive activity of R-carvedilol, the non-β-blocking enantiomer present in the racemic β-blocker, carvedilol. As R-carvedilol does not have β-blocking activity, such a preventive treatment would not lead to common cardiovascular side effects of β-blockers.
Collapse
Affiliation(s)
- Sherry Liang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Md Abdullah Shamim
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Ayaz Shahid
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Mengbing Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Kristan H Cleveland
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Cyrus Parsa
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California.,Department of Pathology, Beverly Hospital, Montebello, California
| | - Robert Orlando
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California.,Department of Pathology, Beverly Hospital, Montebello, California
| | - Bradley T Andresen
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Ying Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California.
| |
Collapse
|
11
|
Dong W, Jiang H, He L, Pan B, Lin L, Song Y, Yang Q. Protein profile of ear auricle cartilage and the important role of ITGB1/PTK2 in microtia. Int J Pediatr Otorhinolaryngol 2020; 137:110235. [PMID: 32896350 DOI: 10.1016/j.ijporl.2020.110235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/27/2020] [Accepted: 06/27/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Microtia is a congenital malformation of the external ear that involves anything from a small reduction in size to a complete absence. The external ear is composed of elastic cartilage which is also the important skeleton of the outer ear. However no previous study explored the difference between abnormal elastic cartilage and normal cartilage in the molecular level. METHODS Microtia cartilage and normal cartilage tissue samples from patients subjected to autologous costal cartilage reconstruction were obtained in surgery. Total proteins were extracted and purified, and then proteomic analyzed via LC-MS/MS using DDA/DIA data collection methods. Proteins were also isolated with lysis beads and then analyzed via antibody chip. Differentially expressed proteins were identified in both experiments and further analyzed with functional enrichment analysis and KEGG pathway analysis. Valuable regulatory gene expression level was verified by RT-PCR. RESULTS A total of 4178 protein types were identified in the DDA experiment. A total of 2154 proteins were quantified, 172 of which were significantly upregulated and 82 downregulated in the microtia group (P < 0.05). Antibody chip detection allowed identification of 584 protein phosphorylation sites with 102 upregulation sites and 9 downregulation sites (P < 0.05). Differentially altered proteins were annotated to 143 KEGG pathways, while differentiated phosphate site-associated genes were annotated into 21 KEGG pathways. Two intersecting pathways, the PI3K/AKT/mTOR pathway and the focal adhesion pathway, may paly important role on ear auricle cartilage development. One item is significant in both differential protein expression and phosphorylation. Integrin beta-1, that is downregulated in protein quantification of the microtia group. The mean ITGB1 mRNA level of the microtia patient group was significantly lower than in the healthy control group (P = 0.0007 < 0.05). And the gene expression of downstream gene PTK2 was also decreased. (P = 0.0288 < 0.05). CONCLUSION The research locates the key protein Integrin Beta-1, and verified it at the mRNA level. The increasing level of ITGB1 and decreasing of PTK2 may play an important role in congenital ear deformity. This research will inspire more otolaryngologists and orthopedics doctors to pay attention to the etiology and mechanism of microtia.
Collapse
Affiliation(s)
- Weiwei Dong
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Haiyue Jiang
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Leren He
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Bo Pan
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Lin Lin
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Yupeng Song
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China
| | - Qinghua Yang
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, China.
| |
Collapse
|
12
|
The β-Blocker Carvedilol Prevented Ultraviolet-Mediated Damage of Murine Epidermal Cells and 3D Human Reconstructed Skin. Int J Mol Sci 2020; 21:ijms21030798. [PMID: 31991834 PMCID: PMC7037760 DOI: 10.3390/ijms21030798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/17/2020] [Accepted: 01/24/2020] [Indexed: 11/17/2022] Open
Abstract
The β-blocker carvedilol prevents ultraviolet (UV)-induced skin cancer, but the mechanism is unknown. Since carvedilol possesses antioxidant activity, this study investigated whether carvedilol prevents oxidative photodamage of skin, a precursor event in skin carcinogenesis. The effects of carvedilol, metoprolol (a β-blocker without antioxidant property), and 4-hydroxycarbazole (4-OHC, a carvedilol synthesis intermediate and a free radical scavenger) were compared on UV- or H2O2-induced cell death and reactive oxygen species (ROS) production in murine epidermal JB6 P+ cells. Although carvedilol attenuated cell death, metoprolol and 4-OHC failed to show protective effects. As expected, increased cellular ROS induced by H2O2 or UV was abolished by carvedilol and 4-OHC, but not by metoprolol. Consistently, carvedilol attenuated the formation of UV-induced cyclobutane pyrimidine dimers (CPDs) and release of prostaglandin E2 in JB6 P+ cells. Carvedilol's activity was further confirmed in full thickness 3D human reconstituted skin, where carvedilol attenuated UV-mediated epidermal thickening, the number of Ki-67 and p53 positive cells as well as CPD formation. Based on pathway-specific Polymerase Chain Reaction (PCR) Array analysis, carvedilol treatment in many cases normalized UV-induced expression changes in DNA repair genes. Thus, carvedilol's photoprotective activity is not attributed to β-blockade or direct ROS-scavenging capacity, but likely via DNA repair regulation.
Collapse
|
13
|
De Giorgi V, Geppetti P, Lupi C, Benemei S. The Role of β-Blockers in Melanoma. J Neuroimmune Pharmacol 2019; 15:17-26. [PMID: 31482435 DOI: 10.1007/s11481-019-09876-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/15/2019] [Indexed: 12/22/2022]
Abstract
Melanoma is one of the most aggressive and less chemotherapy-responsive human cancers, representing a major public health issue worldwide. The early diagnosis still represents the best approach in order to reduce mortality, especially in advanced stages. Preclinical evidence, collected through several in vitro and in vivo models, has been accumulating about the pathophysiological involvement of β-adrenoceptors in melanoma progression. This involvement has been paralleled by the evidence that drugs blocking β-adrenoceptors (β-blockers) may have a relevant role in the treatment of melanoma and in the prevention of its progression. β-blockers are a class of drugs extensively used in clinical practice, not limited to cardiovascular therapeutics. Evidence collected through retrospective and prospective observational studies suggests that treatment with β-blockers, mainly propranolol, is able to delay melanoma progression. Although conclusive evidence is still lacking, current knowledge proposes β-blockers as an opportunity for antitumor treatment in melanoma. Clinical trials are needed in order to prove their claimed efficacy. Graphical Abstract.
Collapse
Affiliation(s)
- Vincenzo De Giorgi
- Division of Dermatology, Department of Surgery and Translational Medicine, University of Florence, Largo Brambilla 3, 50134, Florence, Italy.
| | - Pierangelo Geppetti
- Headache Centre, Careggi University Hospital, Department of Health Sciences, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Chiara Lupi
- Headache Centre, Careggi University Hospital, Department of Health Sciences, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Silvia Benemei
- Headache Centre, Careggi University Hospital, Department of Health Sciences, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| |
Collapse
|
14
|
Cleveland KH, Liang S, Chang A, Huang KM, Chen S, Guo L, Huang Y, Andresen BT. Carvedilol inhibits EGF-mediated JB6 P+ colony formation through a mechanism independent of adrenoceptors. PLoS One 2019; 14:e0217038. [PMID: 31107911 PMCID: PMC6527222 DOI: 10.1371/journal.pone.0217038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/02/2019] [Indexed: 01/13/2023] Open
Abstract
Carvedilol is reported to prevent cancers in humans and animal models. However, a molecular mechanism has yet to be established, and the extent to which other β-blockers are chemopreventive remains relatively unknown. A comparative pharmacological approach was utilized with the expectation that a mechanism of action could be devised. JB6 Cl 41-5a (JB6 P+) murine epidermal cells were used to elucidate the chemopreventative properties of β-blockers, as JB6 P+ cells recapitulate in vivo tumor promotion and chemoprevention. The initial hypothesis was that β-blockers that are GRK/β-arrestin biased agonists, like carvedilol, are chemopreventive. Sixteen β-blockers of different classes, isoproterenol, and HEAT HCl were individually co-administered with epidermal growth factor (EGF) to JB6 P+ cells to examine the chemopreventative properties of each ligand. Cytotoxicity was examined to ensure that the anti-transformation effects of each ligand were not due to cellular growth inhibition. Many of the examined β-blockers suppressed EGF-induced JB6 P+ cell transformation in a non-cytotoxic and concentration-dependent manner. However, the IC50 values are high for the most potent inhibitors (243, 326, and 431 nM for carvedilol, labetalol, and alprenolol, respectively) and there is no correlation between pharmacological properties and inhibition of transformation. Therefore, the role of α1- and β2-adrenergic receptors (AR) was examined by standard competition assays and shRNA targeting β2-ARs, the only β-AR expressed in JB6 P+ cells. The results reveal that pharmacological inhibition of α1- and β2-ARs and genetic knockdown of β2-ARs did not abrogate carvedilol-mediated inhibition of EGF-induced JB6 P+ cell transformation. Furthermore, topical administration of carvedilol protected mice from UV-induced skin damage, while genetic ablation of β2-ARs increased carvedilol-mediated effects. Therefore, the prevailing hypothesis that the chemopreventive property of carvedilol is mediated through β-ARs is not supported by this data.
Collapse
Affiliation(s)
- Kristan H. Cleveland
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, United States of America
| | - Sherry Liang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, United States of America
| | - Andy Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, United States of America
| | - Kevin M. Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, United States of America
| | - Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Ying Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, United States of America
- * E-mail: (YH); (BTA)
| | - Bradley T. Andresen
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, United States of America
- * E-mail: (YH); (BTA)
| |
Collapse
|
15
|
Bond RA, Lucero Garcia-Rojas EY, Hegde A, Walker JKL. Therapeutic Potential of Targeting ß-Arrestin. Front Pharmacol 2019; 10:124. [PMID: 30894814 PMCID: PMC6414794 DOI: 10.3389/fphar.2019.00124] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/31/2019] [Indexed: 12/22/2022] Open
Abstract
ß-arrestins are multifunctional proteins that modulate heptahelical 7 transmembrane receptors, also known as G protein-coupled receptors (GPCRs), a superfamily of receptors that regulate most physiological processes. ß-arrestin modulation of GPCR function includes termination of G protein-dependent signaling, initiation of ß-arrestin-dependent signaling, receptor trafficking to degradative or recycling pathways, receptor transactivation, transcriptional regulation, and localization of second messenger regulators. The pleiotropic influence ß-arrestins exert on these receptors regulates a breadth of physiological functions, and additionally, ß-arrestins are involved in the pathophysiology of numerous and wide-ranging diseases, making them prime therapeutic targets. In this review, we briefly describe the mechanisms by which ß-arrestins regulate GPCR signaling, including the functional cellular mechanisms modulated by ß-arrestins and relate this to observed pathophysiological responses associated with ß-arrestins. We focus on the role for ß-arrestins in transducing cell signaling; a pathway that is complementary to the classical G protein-coupling pathway. The existence of these GPCR dual signaling pathways offers an immense therapeutic opportunity through selective targeting of one signaling pathway over the other. Finally, we will consider several mechanisms by which the potential of dual signaling pathway regulation can be harnessed and the implications for improved disease treatments.
Collapse
Affiliation(s)
- Richard A Bond
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Emilio Y Lucero Garcia-Rojas
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Akhil Hegde
- School of Nursing, Duke University, Durham, NC, United States
| | | |
Collapse
|