51
|
Sanidas E, Velliou M, Papadopoulos D, Fotsali A, Iliopoulos D, Mantzourani M, Toutouzas K, Barbetseas J. Antihypertensive Drugs and Risk of Cancer: Between Scylla and Charybdis. Am J Hypertens 2020; 33:1049-1058. [PMID: 32529212 DOI: 10.1093/ajh/hpaa098] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/21/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
Antihypertensive drugs namely angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, calcium channel blockers, beta blockers, and diuretics are among the most clearly documented regimens worldwide with an overall cardioprotective benefit. Given that malignancy is the second leading cause of mortality, numerous observational studies aimed to investigate the carcinogenic potential of these agents with conflicting results. The purpose of this review was to summarize current data in an effort to explore rare side effects and new mechanisms linking antihypertensive drugs with the risk of developing cancer.
Collapse
Affiliation(s)
- Elias Sanidas
- Hypertension Excellence Centre—ESH, Department of Cardiology, LAIKO General Hospital, Athens, Greece
| | - Maria Velliou
- Hypertension Excellence Centre—ESH, Department of Cardiology, LAIKO General Hospital, Athens, Greece
| | - Dimitrios Papadopoulos
- Hypertension Excellence Centre—ESH, Department of Cardiology, LAIKO General Hospital, Athens, Greece
| | - Anastasia Fotsali
- Hypertension Excellence Centre—ESH, Department of Cardiology, LAIKO General Hospital, Athens, Greece
| | - Dimitrios Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research “N.S. Christeas”, University of Athens, Medical School, Athens, Greece
| | - Marina Mantzourani
- 1st Department of Internal Medicine, LAIKO General Hospital, University of Athens, Medical School, Athens, Greece
| | - Konstantinos Toutouzas
- University of Athens, 1st Department of Cardiology, Hippokrateion Hospital, Athens, Greece
| | - John Barbetseas
- Hypertension Excellence Centre—ESH, Department of Cardiology, LAIKO General Hospital, Athens, Greece
| |
Collapse
|
52
|
Impairment of Hypoxia-Induced CA IX by Beta-Blocker Propranolol-Impact on Progression and Metastatic Potential of Colorectal Cancer Cells. Int J Mol Sci 2020; 21:ijms21228760. [PMID: 33228233 PMCID: PMC7699498 DOI: 10.3390/ijms21228760] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
The coexistence of cancer and other concomitant diseases is very frequent and has substantial implications for treatment decisions and outcomes. Beta-blockers, agents that block the beta-adrenergic receptors, have been related also to cancers. In the model of multicellular spheroids formed by colorectal cancer cells we described a crosstalk between beta-blockade by propranolol and tumour microenvironment. Non-selective beta-blocker propranolol decreased ability of tumour cells to adapt to hypoxia by reducing levels of HIF1α and carbonic anhydrase IX in 3D spheroids. We indicated a double action of propranolol in the tumour microenvironment by inhibiting the stability of HIF1α, thus mediating decrease of CA IX expression and, at the same time, by its possible effect on CA IX activity by decreasing the activity of protein kinase A (PKA). Moreover, the inhibition of β-adrenoreceptors by propranolol enhanced apoptosis, decreased number of mitochondria and lowered the amount of proteins involved in oxidative phosphorylation (V-ATP5A, IV-COX2, III-UQCRC2, II-SDHB, I-NDUFB8). Propranolol reduced metastatic potential, viability and proliferation of colorectal cancer cells cultivated in multicellular spheroids. To choose the right treatment strategy, it is extremely important to know how the treatment of concomitant diseases affects the superior microenvironment that is directly related to the efficiency of anti-cancer therapy
Collapse
|
53
|
β-Adrenoceptor Activation in Breast MCF-10A Cells Induces a Pattern of Catecholamine Production Similar to that of Tumorigenic MCF-7 Cells. Int J Mol Sci 2020; 21:ijms21217968. [PMID: 33120955 PMCID: PMC7672553 DOI: 10.3390/ijms21217968] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/13/2020] [Accepted: 10/25/2020] [Indexed: 12/23/2022] Open
Abstract
Adrenaline, which participates in the neuroendocrine response that occurs during stress and perimenopause, may be tumorigenic. This exploratory study aimed at investigating whether non-tumorigenic and tumorigenic human breast epithelial cell lines are able to synthesize adrenaline. The study was carried out in non-tumorigenic (MCF-10A) and tumorigenic (MCF-7) human breast cell lines. Expression of enzymes involved in adrenaline synthesis was characterized by RT-qPCR, immunocytochemistry and western blot. Catecholamines and analogue compounds were quantified by HPLC-ECD. Functional assessment of the impact of drugs on cells’ tumorigenic potential was assessed by determination of cell viability and clonogenic ability. Both MCF-10A and MCF-7 cells produce catecholamines, but the capacity to produce adrenaline is lower in MCF-10A cells. β-adrenoceptor activation increases the capacity of MCF-10A cells to produce adrenaline and favor both cell viability and colony formation. It is concluded that exposure of human breast epithelial cells to β-adrenoceptor agonists increases cell proliferation and the capacity to produce adrenaline, creating an autocrine potential to spread these adrenergic effects in a feed-forward loop. It is conceivable that these effects are related to tumorigenesis, bringing a new perspective to understand the claimed anticancer effects of propranolol and the increase in breast cancer incidence caused by stress or during perimenopause.
Collapse
|
54
|
Zahalka AH, Fram E, Lin W, Mohn L, Frenette PS, Agalliu I, Watts KL. Use of beta-blocker types and risk of incident prostate cancer in a multiethnic population. Urol Oncol 2020; 38:794.e11-794.e16. [PMID: 32307329 DOI: 10.1016/j.urolonc.2020.03.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/15/2020] [Accepted: 03/24/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE Increased adrenergic innervation is observed in prostate cancer (CaP) and is associated with aggressive disease. Emerging evidence suggests that beta-adrenergic blockade inhibits CaP progression. However, the association between type of beta-blocker use and risk of incident CaP on initial prostate biopsy has not been investigated in multiethnic populations. MATERIALS AND METHODS A retrospective study of racially/ethnically diverse men (64% African-American and Hispanic), who underwent initial prostate biopsy between 2006 and 2016 in a large healthcare system was performed. Oral use of beta-blocker type was assessed by reviewing active prescriptions within the 5-year period preceding initial biopsy. Patient demographics and clinical factors were collected. RESULTS Of 4,607 men who underwent initial prostate biopsy, 4,516 met criteria and 2,128 had a biopsy positive for CaP; 20% high-risk, 41% intermediate-risk, and 39% low or very-low risk (National Comprehensive Cancer Network classification). Overall, 15% of patients were taking a beta-blocker prior to initial biopsy, with Metoprolol, Atenolol, and Carvedilol accounting for the majority. Of beta-blocker types, Atenolol alone was associated with a 38% reduction in odds of incident CaP (P= 0.01), with a 40% and 54% reduction in risks of National Comprehensive Cancer Network intermediate and high-risk CaP (P = 0.03 and P = 0.03, respectively) compared to men not taking a beta-blocker. Furthermore, longer duration of Atenolol use (3-5 years) was associated with a 54% and 72% reduction in intermediate and high-risk disease, (P = 0.03 and P = 0.03, respectively). CONCLUSIONS Among beta blocker types, long-term Atenolol use is associated with a significant reduction in incident CaP risk on initial prostate biopsy for clinically-significant intermediate and high-risk disease compared to men not taking a beta-blocker.
Collapse
Affiliation(s)
- Ali H Zahalka
- Department of Urology, Albert Einstein College of Medicine / Montefiore Medical Center, Bronx, NY; Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Ethan Fram
- Department of Urology, Albert Einstein College of Medicine / Montefiore Medical Center, Bronx, NY
| | - Wilson Lin
- Department of Urology, Albert Einstein College of Medicine / Montefiore Medical Center, Bronx, NY
| | - Larkin Mohn
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Paul S Frenette
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY; Ruth L. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Ilir Agalliu
- Department of Urology, Albert Einstein College of Medicine / Montefiore Medical Center, Bronx, NY; Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Kara L Watts
- Department of Urology, Albert Einstein College of Medicine / Montefiore Medical Center, Bronx, NY
| |
Collapse
|
55
|
Beta-adrenergic blocker inhibits oral carcinogenesis and reduces tumor invasion. Cancer Chemother Pharmacol 2020; 86:681-686. [PMID: 32980903 DOI: 10.1007/s00280-020-04149-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/16/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Beta-adrenergic signaling can influence cancer progression and the use of beta blockers as adjuvant drugs in oncologic patients has been suggested. However, the involvement of beta-adrenergic blockers in tumorigenesis is poorly understood. This study investigated the action of beta-adrenergic blocker propranolol on tumor onset using a preclinical model of chemically induced oral cancer. METHODS Thirty-two male Wistar rats were subjected to daily subcutaneous injection of beta-blocker propranolol (10 mg/kg; SubQ), while another 32 rats received only a PBS injection (sham group). One week after starting propranolol treatment, all rats were submitted to chemical induction of oral carcinogenesis with 4-nitroquinoline-1-oxide (4NQO). After 16 weeks, they were assessed for occurrence of oral squamous cell carcinoma (OSCC), in addition to measurement of tumor volume and thickness, and tissue levels of cytokines IL-6, TNF-alpha and IL-10 in the tumor microenvironment. RESULTS Propranolol treatment reduced the occurrence of OSCC by 31%, 95% CI ( - 127, 216). Beta-adrenergic blocker significantly decreased thickness of OSCC when compared with PBS. Rats treated with propranolol exhibited a lower tumor volume when compared with control rats, but this result did not reach statistical significance. Tumors from propranolol-treated rats exhibited reduced concentrations of pro-inflammatory cytokines IL-6 and TNF-α. There was no difference in the IL-10 levels between tumors from propranolol- and sham-treated rats. CONCLUSION Beta-adrenergic signaling may be one of the mechanisms associated with chemically induced oral carcinogenesis.
Collapse
|
56
|
Renin-Angiotensin-Aldosterone System-based Antihypertensive Agents and the Risk of Colorectal Cancer Among Medicare Beneficiaries. Epidemiology 2020; 30:867-875. [PMID: 31348009 DOI: 10.1097/ede.0000000000001065] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Biologic evidence suggests that angiotensin II may play a role in tumor progression or growth. We compared the short-term colorectal cancer (CRC) risk among initiators of angiotensin-converting enzyme inhibitors (ACEI) or angiotensin receptor blockers (ARB) versus guideline-recommended clinical alternatives (beta blockers, calcium channel blockers [CCB], and thiazides). METHODS We conducted a new-user cohort study on U.S. Medicare beneficiaries aged over 65 years, who initiated antihypertensive monotherapy during 2007-2013 and were free of cancer diagnosis before drug initiation. Follow-up began 6 months postinitiation to allow time for the diagnostic delay. We estimated hazard ratios (HR) with 95% confidence intervals (CI) using propensity score weighted Cox regression, overall and stratified by time since drug initiation, and 5-year cumulative risk differences (RD) using Kaplan-Meier estimator. We assessed the potential for unmeasured confounding using supplemental data from Medicare Current Beneficiary Survey. RESULTS For analyses without censoring for treatment changes, we observed 532 CRC events among 111,533 ACEI/ARB initiators. After a median follow-up of 2.2 years (interquartile range: 1.0-3.7), CRC risk was similar between ACEI/ARB and active comparators, with adjusted HRs of 1.0 (95% CI = 0.85, 1.1) for ACEI/ARB versus beta blockers, 1.2 (95% CI = 0.97, 1.4) for ACEI/ARB versus CCB and 1.0 (95% CI = 0.80, 1.3) for ACEI/ARB versus thiazide. Five-year RDs and as-treated analyses, which censored follow-up at medication changes, produced similar findings. CONCLUSIONS Based on real-world antihypertensive utilization patterns in Medicare beneficiaries, our study suggests no association between ACEI/ARB initiation and the short-term CRC risk.
Collapse
|
57
|
Mohammed MA, Ahmed MT, Anwer BE, Aboshanab KM, Aboulwafa MM. Propranolol, chlorpromazine and diclofenac restore susceptibility of extensively drug-resistant (XDR)-Acinetobacter baumannii to fluoroquinolones. PLoS One 2020; 15:e0238195. [PMID: 32845920 PMCID: PMC7449414 DOI: 10.1371/journal.pone.0238195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Nosocomial infections caused by extensively drug-resistant (XDR) or Pan-Drug resistant (PDR) Acinetobacter (A.) baumannii have recently increased dramatically creating a medical challenge as therapeutic options became very limited. The aim of our study was to investigate the antibiotic-resistance profiles and evaluate the various combinations of ciprofloxacin (CIP) or levofloxacin (LEV) with antimicrobial agents and non-antimicrobial agents to combat antimicrobial resistance of XDR A. baumannii. A total of 100 (6.25%) A. baumannii clinical isolates were recovered from 1600 clinical specimens collected from hospitalized patients of two major university hospitals in Upper Egypt. Antimicrobial susceptibility tests were carried out according to CLSI guidelines. Antimicrobial susceptibility testing of the respective isolates showed a high percentage of bacterial resistance to 19 antimicrobial agents ranging from 76 to99%. However, a lower percentage of resistance was observed for only colistin (5%) and doxycycline (57%). The isolates were categorized as PDR (2; 2%), XDR (68; 68%), and multi-drug resistant (MDR) (30; 30%). Genotypic analysis using ERIC-PCR on 2 PDR and 32 selected XDR isolates showed that they were not clonal. Combinations of CIP or LEV with antibiotics (including, ampicillin, ceftriaxone, amikacin, or doxycycline) were tested on these A. baumannii non-clonal isolates using standard protocols where fractional inhibitory concentrations (-FICs) were calculated. Results of the respective combinations showed synergism in 23.5%, 17.65%, 32.35%, 17.65% and 26.47%, 8.28%, 14.71%, 26.47%, of the tested isolates, respectively. CIP or LEV combinations with either chlorpromazine (CPZ) 200 μg/ml, propranolol (PR) in two concentrations, 0.5 mg/ml and 1.0 mg/ml or diclofenac (DIC) 4 mg/ml were carried out and the MIC decrease factor (MDF) of each isolate was calculated and results showed synergism in 44%, 50%, 100%, 100% and 94%, 85%, 100%, 100%, of the tested isolates, respectively. In conclusion, combinations of CIP or LEV with CPZ, PR, or DIC showed synergism in most of the selected PDR and XDR A. baumannii clinical isolates. However, these combinations have to be re-evaluated in vivo using appropriate animal models infected by XDR- or PDR- A. baumannii.
Collapse
Affiliation(s)
- Mostafa A. Mohammed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Assiut, Egypt
| | - Mohammed T. Ahmed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Assiut, Egypt
| | - Bahaa E. Anwer
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Assiut, Egypt
| | - Khaled M. Aboshanab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohammad M. Aboulwafa
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
58
|
Targeting β2-Adrenergic Receptors Shows Therapeutical Benefits in Clear Cell Renal Cell Carcinoma from Von Hippel-Lindau Disease. J Clin Med 2020; 9:jcm9092740. [PMID: 32854260 PMCID: PMC7563408 DOI: 10.3390/jcm9092740] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Von Hippel–Lindau (VHL), is a rare autosomal dominant inherited cancer in which the lack of VHL protein triggers the development of multisystemic tumors such us retinal hemangioblastomas (HB), CNS-HB, and clear cell renal cell carcinoma (ccRCC). ccRCC ranks third in terms of incidence and first in cause of death. Standard systemic therapies for VHL-ccRCC have shown limited response, with recurrent surgeries being the only effective treatment. Targeting of β2-adrenergic receptor (ADRB) has shown therapeutic antitumor benefits on VHL-retinal HB (clinical trial) and VHL-CNS HB (in vitro). Therefore, the in vitro and in vivo antitumor benefits of propranolol (ADRB-1,2 antagonist) and ICI-118,551 (ADRB-2 antagonist) on VHL−/− ccRCC primary cultures and 786-O tumor cell lines have been addressed. Propranolol and ICI-118,551 activated apoptosis inhibited gene and protein expression of HIF-2α, CAIX, and VEGF, and impaired partially the nuclear internalization of HIF-2α and NFĸB/p65. Moreover, propranolol and ICI-118,551 reduced tumor growth on two in vivo xenografts. Finally, ccRCC patients receiving propranolol as off-label treatment have shown a positive therapeutic response for two years on average. In summary, propranolol and ICI-118,551 have shown antitumor benefits in VHL-derived ccRCC, and since ccRCCs comprise 63% of the total RCCs, targeting ADRB2 becomes a promising drug for VHL and other non-VHL tumors.
Collapse
|
59
|
Abstract
Propranolol is one of the first medications of the beta-blocker used for antihypertensive drugs. This study reports the facile route for the synthesis of propranolol and its novel derivatives. Herein, propranolol synthesis proceeded from 1-naphthol and isopropylamine under mild and less toxic conditions. Novel propranolol derivatives were designed by reactions of propranolol with benzoyl chloride, pyridinium chlorochromate, and n-butyl bromide through esterification, oxidation reduction, and alkylation, respectively. The isolation and purity of compounds were conducted using column chromatography and thin-layer chromatography. Mass spectrometry and 1H-NMR spectroscopy were applied to identify new compounds structure. Propranolol derivatives from 2-chlorobenzoyl chloride (compound 3), 2-fluorobenzoyl chloride (compound 5), and especially acetic anhydride (compound 6) manifested high yields and significantly increased water solubility. Six semisynthetic propranolol derivatives promise to improve antioxidative and biological activities.
Collapse
|
60
|
Down-regulation of ADRB2 expression is associated with small cell neuroendocrine prostate cancer and adverse clinical outcomes in castration-resistant prostate cancer. Urol Oncol 2020; 38:931.e9-931.e16. [PMID: 32624423 DOI: 10.1016/j.urolonc.2020.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/13/2020] [Accepted: 06/02/2020] [Indexed: 01/04/2023]
Abstract
OBJECTIVES The net oncogenic effect of β2-adrenergic receptor ADRB2, whose downstream elements induce neuroendocrine differentiation and whose expression is regulated by EZH2, is unclear. ADRB2 expression and associated clinical outcomes in metastatic castration-resistant prostate cancer (mCRPC) are unknown. METHODS AND MATERIALS This was a retrospective analysis of a multi-center, prospectively enrolled cohort of mCRPC patients. Metastatic biopsies were obtained at progression, and specimens underwent laser capture microdissection and RNA-seq. ADRB2 expression was stratified by histology and clustering based on unsupervised hierarchical transcriptome analysis and correlated with EZH2 expression; an external dataset was used for validation. The association between ADRB2 expression and overall survival (OS) was assessed by log-rank test and a multivariable Cox proportional hazard model. RESULTS One hundred and twenty-seven patients with progressive mCRPC had sufficient metastatic tumor for RNA-seq. ADRB2 expression was lowest in the small cell-enriched transcriptional cluster (P < 0.01) and correlated inversely with EZH2 expression (r = -0.28, P < 0.01). These findings were validated in an external cohort enriched for neuroendocrine differentiation. Patients with tumors harboring low ADRB2 expression (lowest quartile) had a shorter median OS than those with higher (9.5 vs. 20.5 months, P = 0.02). In multivariable analysis, low ADRB2 expression was associated with a trend toward shorter OS (HR for death = 1.54, 95%CI 0.98-2.44). Conversely, higher expression of upstream transcriptional regulator EZH2 was associated with shortened OS (HR for death = 3.01, 95%CI 1.12-8.09). CONCLUSIONS Low ADRB2 expression is associated with neuroendocrine differentiation and is associated with shortened survival. EZH2 is a potential therapeutic target for preventing neuroendocrine transdifferentiation and improving outcomes in mCRPC. Further studies of agents targeting β-adrenergic signaling are warranted.
Collapse
|
61
|
Preventing Lethal Prostate Cancer with Diet, Supplements, and Rx: Heart Healthy Continues to Be Prostate Healthy and "First Do No Harm" Part III. Curr Urol Rep 2020; 21:22. [PMID: 32367257 DOI: 10.1007/s11934-020-00972-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW To discuss the overall and latest observations of the effect of diet, lifestyle, supplements, preventive vaccinations, and some prescription heart healthy medications for prostate cancer prevention within a 3-part series of publications. RECENT FINDINGS The concept of maximizing heart health to prevent aggressive prostate cancer continues to be solidified with additional prospective observational and randomized controlled trial data. Heart healthy is prostate healthy, but heart unhealthy is prostate unhealthy. The primary goal for medical providers of reducing all-cause and cardiovascular disease (CVD) morbidity and mortality correlates with maximizing prostate cancer prevention. The obesity epidemic in children and adults along with research from multiple, diverse disciplines has only strengthened the nexus between heart and prostate health. Greater dietary adherence toward a variety of healthy foods is associated with a graded reduction in the probability of CVD and aggressive cancer. Preventing prostate cancer via dietary supplements should encourage a "first do no harm" or less-is-more approach until future evidence can reverse the concerning trend that more supplementation has resulted in either no impact or an increased risk of prostate cancer. Supplements to reduce side effects of some cancer treatments appear to have more encouraging data. A discussion of quality control (QC) before utilizing any pill also requires attention. Medications or interventions that potentially improve heart health including statins, aspirin, and metformin (S.A.M.), specific beta-blocker medications, and even preventive vaccines are in general generic, low cost, "natural," and should continue to garner research interest. A watershed moment in medical education has arrived where the past perception of a diverse number of trees seemingly separated by vast distances, in reality, now appears to exist within the same forest.
Collapse
|
62
|
Baccino D, Merlo G, Cozzani E, Rosa GM, Tini G, Burlando M, Parodi A. Cutaneous effects of antihypertensive drugs. GIORN ITAL DERMAT V 2020; 155:202-211. [DOI: 10.23736/s0392-0488.19.06360-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
63
|
Xie Y, Xu P, Wang M, Zheng Y, Tian T, Yang S, Deng Y, Wu Y, Zhai Z, Hao Q, Song D, Zhang D, Dai Z. Antihypertensive medications are associated with the risk of kidney and bladder cancer: a systematic review and meta-analysis. Aging (Albany NY) 2020; 12:1545-1562. [PMID: 31968309 PMCID: PMC7053620 DOI: 10.18632/aging.102699] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/26/2019] [Indexed: 12/24/2022]
Abstract
Several studies have indicated that the use of antihypertensive medications may influence the incidence of bladder/kidney cancer, with some scholars refuting any such association. Hence, a systematic review is needed to verify this linkage. we comprehensively searched PubMed, Embase, Web of Science, and the Cochrane Library for original studies reporting a relationship between antihypertensive medications and risk of bladder/kidney cancer. We included 31 articles comprising 3,352,264 participants. We found a significant association between the risk of kidney cancer and any antihypertensive medications use (relative risk (RR) = 1.45, 95% CI 1.20-1.75), as well as angiotensin-converting enzyme inhibitors (RR = 1.24, 95% CI 1.04-1.48), angiotensin II receptor blockers (ARB) (RR = 1.29, 95% CI:1.22-1.37), beta-blockers (RR = 1.36, 95% CI 1.11-1.66), calcium-channel blockers (RR = 1.65, 95% CI 1.54-1.78) and diuretics (RR = 1.34, 95% CI 1.19-1.51). In case of bladder cancer, a statistical significance was observed with the use of ARB (RR = 1.07, 95% CI 1.03-1.11) but not with the other antihypertensive medications. There was a linear association between the duration of antihypertensive medications and the risk of kidney cancer (P = 0.061 for a non-linear trend) and the pooled RR for the per year increase in antihypertensive medications duration of use was 1.02 (95% CI: 1.01-1.02). Our results indicate that there is a significant association between each class of antihypertensive medications and the risk of kidney cancer, and this trend presented as a positive linear association. Furthermore, the use of ARB has been linked to the risk of bladder cancer.
Collapse
Affiliation(s)
- Yuxiu Xie
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Xu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zheng
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tian Tian
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Si Yang
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yujiao Deng
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Wu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhen Zhai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dingli Song
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dai Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
64
|
The Role of Non-Selective β-Blockers in Compensated Cirrhotic Patients without Major Complications. ACTA ACUST UNITED AC 2019; 56:medicina56010014. [PMID: 31905956 PMCID: PMC7022668 DOI: 10.3390/medicina56010014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/22/2019] [Accepted: 12/22/2019] [Indexed: 12/17/2022]
Abstract
Background and Objectives: Non-selective β-blockers (NSBB) could prevent decompensation and hepatocellular carcinoma (HCC) in cirrhotic patients with clinically significant portal hypertension (CSPH), but remained uncertain for compensated cirrhotic patients without major complications. We aimed to compare the clinical outcomes between propranolol users and non-users of a CC group without major complications. Material and Methods: We conducted this population-based cohort study by using the Taiwanese Longitudinal Health Insurance Database 2000. Propranolol users (classified as cumulative defined daily dose (cDDD)) and non-PPL users were matched with a 1:1 propensity score in both cohorts. Results: This study comprised 6896 propranolol users and 6896 non-propranolol users. There was no significant impact on the development of spontaneous bacterial peritonitis between the two groups (aHR: 1.24, 95% confidence interval (CI): 0.88~1.75; p = 0.2111). Male gender, aged condition, and non-liver related diseases (peripheral vascular disease, cerebrovascular disease, dementia, pulmonary disease, and renal disease) were the independent risk factors of mortality. PPL users had significantly lower incidence of HCC development than non-users (aHR: 0.81, p = 0.0580; aHR: 0.80, p = 0.1588; and aHR: 0.49, p < 0.0001 in the groups of 1–28, 29–90, and >90 cDDD, respectively). Conclusion: The current study suggested that high cumulative doses of propranolol could decrease the risk of hepatocellular carcinoma among compensated cirrhotic patients without major complications. Further large-scale prospective studies are still required to confirm the findings in this study. Results: It remained uncertain whether non-selective β-blockers (NSBB) could prevent decompensation and hepatocellular carcinoma (HCC) in compensatory cirrhotic patients without major complications. This study aimed to compare the clinical outcomes between propranolol users and non-users of the CC group without major complications.
Collapse
|
65
|
β-Adrenergic Signaling in Lung Cancer: A Potential Role for Beta-Blockers. J Neuroimmune Pharmacol 2019; 15:27-36. [PMID: 31828732 DOI: 10.1007/s11481-019-09891-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/29/2019] [Indexed: 12/28/2022]
Abstract
Lung cancer results in more patient deaths each year than any other cancer type. Additional treatment strategies are needed to improve clinical responses to approved treatment modalities and prevent the emergence of resistant disease. Catecholamines including norepinephrine and epinephrine are elevated as a result of chronic stress and mediate their physiological effects through activation of adrenergic receptors on target tissues. Lung cancer cells express β-adrenergic receptors (β-ARs), and numerous preclinical studies indicate that β2-AR signaling on lung cancer cells facilities cellular programs including proliferation, motility, apoptosis resistance, epithelial-to-mesenchymal transition, metastasis, and the acquisition of an angiogenic and immunosuppressive phenotype. Here, we review the preclinical and clinical evidence supporting a potential role for beta-blockers in improving the clinical outcome of lung cancer patients. Graphical Abstract Catecholamines including norepinephrine and epinephrine act of β-ARs expressed on NSCLC tumor cells and activate pathways regulating tumor progression.
Collapse
|
66
|
Tan DC, Roth IM, Wickremesekera AC, Davis PF, Kaye AH, Mantamadiotis T, Stylli SS, Tan ST. Therapeutic Targeting of Cancer Stem Cells in Human Glioblastoma by Manipulating the Renin-Angiotensin System. Cells 2019; 8:cells8111364. [PMID: 31683669 PMCID: PMC6912312 DOI: 10.3390/cells8111364] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with glioblastoma (GB), a highly aggressive brain tumor, have a median survival of 14.6 months following neurosurgical resection and adjuvant chemoradiotherapy. Quiescent GB cancer stem cells (CSCs) invariably cause local recurrence. These GB CSCs can be identified by embryonic stem cell markers, express components of the renin-angiotensin system (RAS) and are associated with circulating CSCs. Despite the presence of circulating CSCs, GB patients rarely develop distant metastasis outside the central nervous system. This paper reviews the current literature on GB growth inhibition in relation to CSCs, circulating CSCs, the RAS and the novel therapeutic approach by repurposing drugs that target the RAS to improve overall symptom-free survival and maintain quality of life.
Collapse
Affiliation(s)
- David Ch Tan
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
| | - Imogen M Roth
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Agadha C Wickremesekera
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Paul F Davis
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem 91120, Israel.
| | - Theo Mantamadiotis
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt 5040, New Zealand.
| |
Collapse
|
67
|
Shepard MJ, Bugarini A, Edwards NA, Lu J, Zhang Q, Wu T, Zhuang Z, Chittiboina P. Repurposing propranolol as an antitumor agent in von Hippel-Lindau disease. J Neurosurg 2019; 131:1106-1114. [PMID: 30497198 PMCID: PMC7265978 DOI: 10.3171/2018.5.jns172879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/10/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Von Hippel-Lindau disease (VHL) is a tumor predisposition syndrome characterized by CNS hemangioblastomas (HBs) and clear cell renal cell carcinomas (RCCs) due to hypoxia-inducible factor activation (pseudohypoxia). Because of the lack of effective medical therapies for VHL, HBs and RCCs account for significant morbidity and mortality, ultimately necessitating numerous neurological and renal surgeries. Propranolol is an FDA-approved pan-beta adrenergic antagonist with antitumor effects against infantile hemangiomas (IHs) and possibly VHL HBs. Here, the authors investigated the antitumor efficacy of propranolol against pseudohypoxia-driven VHL-HBs and VHL-RCCs. METHODS Patient-derived VHL-associated HBs (VHL-HBs) or 786-O-VHL-/- RCC cells were treated with clinically relevant concentrations of propranolol in vitro and assessed with viability assays, flow cytometry, quantitative real-time polymerase chain reaction, and western blotting. In vivo confirmation of propranolol antitumor activity was confirmed in athymic nude mice bearing 786-O xenograft tumors. Lastly, patients enrolled in a VHL natural history study (NCT00005902) were analyzed for incidental propranolol intake. Propranolol activity against VHL-HBs was assessed retrospectively with volumetric HB growth kinetic analysis. RESULTS Propranolol decreased HB and RCC viability in vitro with IC50 (half maximal inhibitory concentration) values of 50 µM and 200 µM, respectively. Similar to prior reports in infantile hemangiomas, propranolol induced apoptosis and paradoxically increased VEGF-A mRNA expression in patient-derived VHL-HBs and 786-O cells. While intracellular VEGF protein levels were not affected by propranolol treatment, propranolol decreased HIF expression in 786-O cells (7.6-fold reduction, p < 0.005). Propranolol attenuated tumor progression compared with control (33% volume reduction at 7 days, p < 0.005) in 786-O xenografted tumor-bearing mice. Three patients (harboring 25 growing CNS HBs) started propranolol therapy during the longitudinal VHL-HB study. HBs in these patients tended to grow slower (median growth rate 27.1 mm3/year vs 13.3 mm3/year) during propranolol treatment (p < 0.0004). CONCLUSIONS Propranolol decreases VHL-HB and VHL-related RCC viability in vitro likely by modulation of VEGF expression and by inducing apoptosis. Propranolol abrogates 786-O xenograft tumor progression in vivo, and retrospective clinical data suggest that propranolol curtails HB growth. These results suggest that propranolol may play a role in the treatment of VHL-related tumors.
Collapse
Affiliation(s)
- Matthew J. Shepard
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Alejandro Bugarini
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Nancy A. Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Jie Lu
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Qi Zhang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda
| | - Tianxia Wu
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
68
|
Abstract
Introduction: Smoking is the main preventable cause of lung cancer. This review summarizes preclinical and clinical data on the mechanisms of smoking-associated cancer development of the major histological lung cancer types small cell lung carcinoma squamous cell carcinoma and pulmonary adenocarcinoma (PAC) and the impact of several factors other than smoking on this process. Areas covered: The role of intracellular signaling induced by nicotinic receptors and beta-adrenergic receptors, the resulting increase in intracellular cyclic adenosine monophosphate (cAMP) as a key driver of PAC and the promoting effects of respiratory tract diseases and their therapeutics, psychological stress and global warming. Expert opinion: Smoking has deleterious effects on the regulation of lung epithelia by neurotransmitter receptors that are further enhanced by gene mutations. Sensitization of the alpha-7 nicotinic receptor (α7nAChR) by COPD enhances the carcinogenic effects of smoking and turns nicotine into a carcinogen. Nicotine vaping may, therefore, cause cancer in individuals with chronic obstructive pulmonary disease. The opposing effects of cAMP on the major lung cancer types indicate that patients with PAC of Clara cell phenotype (PAC-Cl) will benefit from treatment with cAMP reducers and suggest that global warming-induced respiratory tract diseases and their therapeutics cause the global increase in the incidence of PAC.
Collapse
Affiliation(s)
- Hildegard M Schuller
- a Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee , Knoxville , TN , USA
| |
Collapse
|
69
|
Overman J, Fontaine F, Wylie-Sears J, Moustaqil M, Huang L, Meurer M, Chiang IK, Lesieur E, Patel J, Zuegg J, Pasquier E, Sierecki E, Gambin Y, Hamdan M, Khosrotehrani K, Andelfinger G, Bischoff J, Francois M. R-propranolol is a small molecule inhibitor of the SOX18 transcription factor in a rare vascular syndrome and hemangioma. eLife 2019; 8:43026. [PMID: 31358114 PMCID: PMC6667216 DOI: 10.7554/elife.43026] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 05/15/2019] [Indexed: 12/15/2022] Open
Abstract
Propranolol is an approved non-selective β-adrenergic blocker that is first line therapy for infantile hemangioma. Despite the clinical benefit of propranolol therapy in hemangioma, the mechanistic understanding of what drives this outcome is limited. Here, we report successful treatment of pericardial edema with propranolol in a patient with Hypotrichosis-Lymphedema-Telangiectasia and Renal (HLTRS) syndrome, caused by a mutation in SOX18. Using a mouse pre-clinical model of HLTRS, we show that propranolol treatment rescues its corneal neo-vascularisation phenotype. Dissection of the molecular mechanism identified the R(+)-propranolol enantiomer as a small molecule inhibitor of the SOX18 transcription factor, independent of any anti-adrenergic effect. Lastly, in a patient-derived in vitro model of infantile hemangioma and pre-clinical model of HLTRS we demonstrate the therapeutic potential of the R(+) enantiomer. Our work emphasizes the importance of SOX18 etiological role in vascular neoplasms, and suggests R(+)-propranolol repurposing to numerous indications ranging from vascular diseases to metastatic cancer.
Collapse
Affiliation(s)
- Jeroen Overman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Frank Fontaine
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jill Wylie-Sears
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Mehdi Moustaqil
- Single Molecule Science, Lowy Cancer Research Centre, The University of New South Wales, Sydney, Australia
| | - Lan Huang
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Marie Meurer
- Centre de Recherche en Cancérologie de Marseille (CRCM Marseille Cancer Research Centre), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University UM105, Marseille, France
| | - Ivy Kim Chiang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Emmanuelle Lesieur
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jatin Patel
- Translational Research Institute, Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Johannes Zuegg
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Eddy Pasquier
- Centre de Recherche en Cancérologie de Marseille (CRCM Marseille Cancer Research Centre), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University UM105, Marseille, France
| | - Emma Sierecki
- Single Molecule Science, Lowy Cancer Research Centre, The University of New South Wales, Sydney, Australia
| | - Yann Gambin
- Single Molecule Science, Lowy Cancer Research Centre, The University of New South Wales, Sydney, Australia
| | | | - Kiarash Khosrotehrani
- Translational Research Institute, Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Gregor Andelfinger
- Department of Pediatrics, University of Montreal, Ste-Justine University Hospital Centre, Montréal, Canada
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Mathias Francois
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| |
Collapse
|
70
|
Hanns P, Paczulla AM, Medinger M, Konantz M, Lengerke C. Stress and catecholamines modulate the bone marrow microenvironment to promote tumorigenesis. Cell Stress 2019; 3:221-235. [PMID: 31338489 PMCID: PMC6612892 DOI: 10.15698/cst2019.07.192] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
High vascularization and locally secreted factors make the bone marrow (BM) microenvironment particularly hospitable for tumor cells and bones to a preferred metastatic site for disseminated cancer cells of different origins. Cancer cell homing and proliferation in the BM are amongst other regulated by complex interactions with BM niche cells (e.g. osteoblasts, endothelial cells and mesenchymal stromal cells (MSCs)), resident hematopoietic stem and progenitor cells (HSPCs) and pro-angiogenic cytokines leading to enhanced BM microvessel densities during malignant progression. Stress and catecholamine neurotransmitters released in response to activation of the sympathetic nervous system (SNS) reportedly modulate various BM cells and may thereby influence cancer progression. Here we review the role of catecholamines during tumorigenesis with particular focus on pro-tumorigenic effects mediated by the BM niche.
Collapse
Affiliation(s)
- Pauline Hanns
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Anna M Paczulla
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Michael Medinger
- Division of Clinical Hematology, University Hospital Basel, Basel, Switzerland
| | - Martina Konantz
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Claudia Lengerke
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland.,Division of Clinical Hematology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
71
|
Kao LT, Huang CC, Lin HC, Huang CY. Antiarrhythmic drug usage and prostate cancer: a population-based cohort study. Asian J Androl 2019; 20:37-42. [PMID: 28857052 PMCID: PMC5753552 DOI: 10.4103/aja.aja_26_17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Even though the relationship between antiarrhythmic drug usage and subsequent prostate cancer (PCa) risk has recently been highlighted, relevant findings in the previous literature are still inconsistent. In addition, very few studies have attempted to investigate the association between sodium channel blockers or potassium channel blockers for arrhythmia and the subsequent PCa risk. Therefore, this cohort study aimed to find the relationship between antiarrhythmic drug usage and the subsequent PCa risk using a population-based dataset. The data used in this study were derived from the Longitudinal Health Insurance Database 2005, Taiwan, China. We respectively identified 9988 sodium channel blocker users, 3663 potassium channel blocker users, 65 966 beta-blocker users, 23 366 calcium channel blockers users, and 7031 digoxin users as the study cohorts. The matched comparison cohorts (one comparison subject for each antiarrhythmic drug user) were selected from the same dataset. Each patient was tracked for a 5-year period to define those who were subsequently diagnosed with PCa. After adjusting for sociodemographic characteristics, comorbidities, and age, Cox proportional hazard regressions found that the hazard ratio (HR) of subsequent PCa for sodium channel blocker users was 1.12 (95% confidence interval [CI]: 0.84–1.50), for potassium channel blocker users was 0.89 (95% CI: 0.59–1.34), for beta-blocker users was 1.08 (95% CI: 0.96–1.22), for calcium channel blocker users was 1.14 (95% CI: 0.95–1.36), and for digoxin users was 0.89 (95% CI: 0.67–1.18), compared to their matched nonusers. We concluded that there were no statistical associations between different types of antiarrhythmic drug usage and subsequent PCa risk.
Collapse
Affiliation(s)
- Li-Ting Kao
- Graduate Institute of Life Science, National Defense Medical Center, Taipei 110, Taiwan, China.,Sleep Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan, China
| | - Chung-Chien Huang
- School of Health Care Administration, Taipei Medical University, Taipei 110, Taiwan, China
| | - Herng-Ching Lin
- Sleep Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan, China.,School of Health Care Administration, Taipei Medical University, Taipei 110, Taiwan, China
| | - Chao-Yuan Huang
- Department of Urology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 110, Taiwan, China.,Department of Urology, National Taiwan University Hospital, Hsin Chu Branch, Hsin Chu City 100, Taiwan, China.,School of Public Health, Taipei Medical University, Taipei 100, Taiwan, China
| |
Collapse
|
72
|
Kim MS, Han KD, Kwon SY. Pre-diagnostic beta-blocker use and head- and neck cancer risk: A nationwide population-based case-control study. Medicine (Baltimore) 2019; 98:e16047. [PMID: 31192963 PMCID: PMC6587613 DOI: 10.1097/md.0000000000016047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
β-blockers have been reported to exhibit potential anticancer effects in various cancer studies. However, few clinical studies concerning head and neck cancer have been conducted. We hypothesized that β-blockers could decrease the incidence of head and neck cancer. Therefore, we investigated the association between β-blocker treatment and head and neck cancer incidence.Between January 2006 and December 2015, we selected 12,127 patients with head and neck cancer for this nationwide study using data from the Korean Health Insurance Review and Assessment Service. The patients were matched 1:5 with 60,635 control participants according to age, sex, and, region. Logistic regression analysis was used to estimate the odds ratios (ORs) and 95% confidence intervals (CIs) of cancer associated with β-blocker treatment. In the analysis, a crude (simple), adjusted model (adjusted model for age, sex, income, region of residence, hypertension, diabetes, and hyperlipidemia) was used.The OR for head and neck cancer incidence was not lower in the β-blocker cohort (OR: 1.18; 95% CI: 1.105-1.26), especially for the oral cavity (OR: 1.165; 95% CI: 1.013-1.340), hypopharynx (OR: 1.555; 95% CI: 1.232-1.963), nasopharynx (OR: 1.251; 95% CI: 1-1.564), and paranasal sinus (OR: 1.378; 95% CI: 1.027-1.849). The duration of β-blocker use was not related to head and neck cancer incidence.This study did not provide evidence that β-blockers can decrease the risk of head and neck cancer.
Collapse
Affiliation(s)
- Min-Su Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam
| | - Kyung Do Han
- Department of Biostatistics, College of Medicine, The Catholic University of Korea
| | - Soon Young Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
73
|
Chang PY, Chung CH, Chang WC, Lin CS, Lin HH, Dai MS, Ho CL, Chien WC. The effect of propranolol on the prognosis of hepatocellular carcinoma: A nationwide population-based study. PLoS One 2019; 14:e0216828. [PMID: 31125347 PMCID: PMC6534323 DOI: 10.1371/journal.pone.0216828] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/29/2019] [Indexed: 12/25/2022] Open
Abstract
Background Beta-blockers can reduce recurrence, metastasis, and mortality in various cancers. In this study, we investigated the effect of propranolol, a non-selective beta-blocker on overall survival (OS) in unresectable/metastatic hepatocellular carcinoma (HCC) and on recurrence-free survival (RFS) in resectable, curable HCC. Methods Data were retrieved from the Taiwan National Health Insurance Research Database between January 2000 and December 2013. Propranolol users (for >1 year) and non-propranolol users were matched using a 1:2 propensity score in both cohorts. Results The unresectable/metastatic HCC cohort comprised 1,560 propranolol users and 3,120 non-propranolol users (control group). On multivariate Cox regression analysis of HCC mortality, propranolol significantly reduced the mortality risk by 22% (hazard ratio [HR] = 0.78, 95% confidence interval [CI] 0.72–0.84, P <0.001). On stratified Cox regression analysis, propranolol also reduced the mortality risk in HCC patients with hepatitis B (HR = 0.92, 95% CI 0.85–0.99, P = 0.045), hepatitis C (HR = 0.85, 95% CI = 0.78–0.92, P = 0.001), liver cirrhosis (HR = 0.78, 95% CI = 0.72–0.85, P <0.001), and diabetes mellitus (HR = 0.87, 95% CI = 0.81–0.94, P = 0.008). The resectable, curable HCC cohort comprised 289 propranolol users and 578 non-propranolol users (control group), but there was no significant difference in RFS (P = 0.762) between propranolol and non-propranolol users. Conclusion This study revealed that propranolol could improve OS in unresectable/metastatic HCC.
Collapse
Affiliation(s)
- Ping-Ying Chang
- Division of Hematology/Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chi-Hsiang Chung
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Wei-Chou Chang
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Hsuan-Hwai Lin
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Ming-Shen Dai
- Division of Hematology/Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Ching-Liang Ho
- Division of Hematology/Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Wu-Chien Chien
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
74
|
Kulik G. ADRB2-Targeting Therapies for Prostate Cancer. Cancers (Basel) 2019; 11:E358. [PMID: 30871232 PMCID: PMC6468358 DOI: 10.3390/cancers11030358] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 01/01/2023] Open
Abstract
There is accumulating evidence that β-2 adrenergic receptor (ADRB2) signaling contributes to the progression and therapy resistance of prostate cancer, whereas availability of clinically tested β-blocker propranolol makes this pathway especially attractive as potential therapeutic target. Yet even in tumors with active ADRB2 signaling propranolol may be ineffective. Inhibition of apoptosis is one of the major mechanisms by which activation of ADRB2 contributes to prostate cancer pathophysiology. The signaling network that controls apoptosis in prostate tumors is highly redundant, with several signaling pathways targeting a few critical apoptosis regulatory molecules. Therefore, a comprehensive analysis of ADRB2 signaling in the context of other signaling mechanisms is necessary to identify patients who will benefit from propranolol therapy. This review discusses how information on the antiapoptotic mechanisms activated by ADRB2 can guide clinical trials of ADRB2 antagonist propranolol as potential life-extending therapy for prostate cancer. To select patients for clinical trials of propranolol three classes of biomarkers are proposed. First, biomarkers of ADRB2/cAMP-dependent protein kinase (PKA) pathway activation; second, biomarkers that inform about activation of other signaling pathways unrelated to ADRB2; third, apoptosis regulatory molecules controlled by ADRB2 signaling and other survival signaling pathways.
Collapse
Affiliation(s)
- George Kulik
- Department of Cancer Biology, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157, USA.
- Department of Life Sciences, Alfaisal University, Riyadh 11533, Saudi Arabia.
| |
Collapse
|
75
|
Di Francesco S, Robuffo I, Caruso M, Giambuzzi G, Ferri D, Militello A, Toniato E. Metabolic Alterations, Aggressive Hormone-Naïve Prostate Cancer and Cardiovascular Disease: A Complex Relationship. ACTA ACUST UNITED AC 2019; 55:medicina55030062. [PMID: 30866568 PMCID: PMC6473682 DOI: 10.3390/medicina55030062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/30/2019] [Accepted: 02/27/2019] [Indexed: 02/06/2023]
Abstract
Background: Epidemiological studies suggest a possible relationship between metabolic alterations, cardiovascular disease and aggressive prostate cancer, however, no clear consensus has been reached. Objective: The aim of the study was to analyze the recent literature and summarize our experience on the association between metabolic disorders, aggressive hormone-naïve prostate cancer and cardiovascular disease. Method: We identified relevant papers by searching in electronic databases such as Scopus, Life Science Journals, and Index Medicus/Medline. Moreover, we showed our experience on the reciprocal relationship between metabolic alterations and aggressive prostate cancer, without the influence of hormone therapy, as well the role of coronary and carotid vasculopathy in advanced prostate carcinoma. Results: Prostate cancer cells have an altered metabolic homeostatic control linked to an increased aggressivity and cancer mortality. The absence of discrimination of risk factors as obesity, systemic arterial hypertension, diabetes mellitus, dyslipidemia and inaccurate selection of vascular diseases as coronary and carotid damage at initial diagnosis of prostate cancer could explain the opposite results in the literature. Systemic inflammation and oxidative stress associated with metabolic alterations and cardiovascular disease can also contribute to prostate cancer progression and increased tumor aggressivity. Conclusions: Metabolic alterations and cardiovascular disease influence aggressive and metastatic prostate cancer. Therefore, a careful evaluation of obesity, diabetes mellitus, dyslipidemia, systemic arterial hypertension, together with a careful evaluation of cardiovascular status, in particular coronary and carotid vascular disease, should be carried out after an initial diagnosis of prostatic carcinoma.
Collapse
Affiliation(s)
- Simona Di Francesco
- Department of Medical and Oral Sciences and Biotechnologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy.
- Department of Urological, Biomedical and Translational Sciences, Federiciana University, 87100 Cosenza, Italy.
| | - Iole Robuffo
- Institute of Molecular Genetics, National Research Council, Section of Chieti, 66100 Chieti, Italy.
| | - Marika Caruso
- Department of Medical and Oral Sciences and Biotechnologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy.
- Department of Urological, Biomedical and Translational Sciences, Federiciana University, 87100 Cosenza, Italy.
| | - Giulia Giambuzzi
- Department of Medical and Oral Sciences and Biotechnologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy.
| | - Deborah Ferri
- Department of Medical and Oral Sciences and Biotechnologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy.
| | - Andrea Militello
- Department of Urological, Biomedical and Translational Sciences, Federiciana University, 87100 Cosenza, Italy.
- Urology and Andrology Section, Villa Immacolata Hospital, 01100 Viterbo, Italy.
| | - Elena Toniato
- Department of Medical and Oral Sciences and Biotechnologies, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy.
| |
Collapse
|
76
|
Siltari A, Murtola TJ, Talala K, Taari K, Tammela TLJ, Auvinen A. Antihypertensive drugs and prostate cancer risk in a Finnish population-based cohort. Scand J Urol 2019; 52:321-327. [PMID: 30698056 DOI: 10.1080/21681805.2018.1559882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The etiology of prostate cancer (PCa) involves environmental and genetic factors. Understanding the role of medication use on PCa risk may clarify the pathophysiological changes and mechanisms in development of cancer. METHODS This study investigated PCa risk in relation to overall use of anti-hypertensive drugs and those with specific mechanisms of action. The study cohort (78,615 men) was linked to the prescription database to obtain information on medication use during 20-year follow-up. Information was obtained on PCa diagnoses, causes of deaths, and for a sub-set on B.M.I. and use of non-prescription drugs. Time-dependent drug use variables hazard ratios (HR) with 95% confidence intervals (CI) were calculated using Cox regression analyses. RESULTS Use of antihypertensive drugs slightly increased PCa risk (HR = 1.16, 95% CI = 1.11-1.22). The risk increase was clearest for metastatic PCa (HR = 1.36, 95% CI = 1.14-1.62). ACE inhibitors, beta-blockers, and diuretics were all separately associated with a small excess risk (HR = 1.10, 95% CI = 1.01-1.19, HR = 1.14, 95% CI = 1.06-1.21, and HR = 1.16, 95% CI = 1.07-1.27, respectively). None of the other groups showed a clear association with PCa risk. CONCLUSIONS The use of antihypertensive drugs was associated with increased prostate cancer risk. Similar risk association for multiple drug groups suggests that the findings may not reflect a direct medication effect, but may be due to underlying hypertension.
Collapse
Affiliation(s)
- Aino Siltari
- a Faculty of Medicine, Pharmacology , University of Helsinki , Helsinki , Finland.,b Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Teemu J Murtola
- b Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland.,c Department of Urology , Tampere University Hospital , Tampere , Finland.,d Department of Surgery , Seinäjoki Central Hospital , Seinäjoki , Finland
| | | | - Kimmo Taari
- f Department of Urology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Teuvo L J Tammela
- b Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland.,c Department of Urology , Tampere University Hospital , Tampere , Finland
| | - Anssi Auvinen
- g Faculty of Social Sciences , University of Tampere , Tampere , Finland
| |
Collapse
|
77
|
Abstract
During cancer progression, tumor cells interact with the neighboring environment, including neuronal tissue. The important influence of the nervous system on growth and metastasis of cancer is now widely accepted. As such, using medications that traditionally target the nervous system may be an avenue toward treating cancer. The focus of this review is to detail how several classes of medications, traditionally used to treat nervous system disorders, impact cancer. Specifically, we review the preclinical and clinical evidence that support the use of anti-β-adrenergic, anticholinergic, antipsychotic, and antidepressant medications to treat some cancers. In addition, we discuss the use of ablative modalities, such as physical and chemical denervation, to treat cancer or protect against cancer development. Using the medications that target the nervous system to treat cancer is a promising addition to an existing therapy or an alternative treatment strategy. Furthermore, rapidly expanding basic science research in this area will likely yield novel cancer therapies that work by targeting the nervous system.
Collapse
|
78
|
Anti-tumor effects of propranolol: Adjuvant activity on a transplanted murine breast cancer model. Biomed Pharmacother 2018; 104:45-51. [PMID: 29758415 DOI: 10.1016/j.biopha.2018.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 11/21/2022] Open
Abstract
Propranolol (Pro), a non-specific β-adrenergic blocking drug, competitively prevents the binding of catecholamines to receptors and suppresses cancer cells. The anti-tumor activity of propranolol has been proved in different kinds of cancers. In this study, we assessed the adjuvant activity of propranolol combined with a tumor vaccine model on the immunological parameters of breast tumor-bearing mice. Breast tumor pieces were implanted into the flank of inbred BALB/C female mice from stock mice. Tumor-bearing mice were treated with tumor antigen lysate vaccine and propranolol/Vaccine (Pro/Vac) combination (as treatment groups), propranolol and PBS (as control groups) for 5 consecutive days, every 12 h. Moreover, all experimental groups received vaccine for three times with one-week interval via s.c injection. After immunization courses, spleens of tumor-bearing mice were removed and dissected, cell suspension was stimulated in vitro, and the cytokine levels in supernatant of splenocytes were measured via commercial ELISA kits. Compared with the vaccine group, immunization with tumor lysate in combination with propranolol significantly increased IL-2, IL-4, IL-12, IL-17, and IFN-γ cytokines. Considering the suppression of tumor growth, propranolol seems to be a potent immunomodulator capable of inducing cellular immune responses against breast cancer.
Collapse
|
79
|
Propranolol sensitizes prostate cancer cells to glucose metabolism inhibition and prevents cancer progression. Sci Rep 2018; 8:7050. [PMID: 29728578 PMCID: PMC5935740 DOI: 10.1038/s41598-018-25340-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/18/2018] [Indexed: 02/04/2023] Open
Abstract
Propranolol, a widely used non-selective beta-adrenergic receptor blocker, was recently shown to display anticancer properties. Its potential to synergize with certain drugs has been also outlined. However, it is necessary to take into account all the properties of propranolol to select a drug that could be efficiently combined with. Propranolol was reported to block the late phase of autophagy. Hence, we hypothesized that in condition enhancing autophagy flux, cancer cells should be especially sensitive to propranolol. 2DG, a glycolysis inhibitor, is an anti-tumor agent having limited effect in monotherapy notably due to induction of pro-survival autophagy. Here, we report that treatment of cancer cells with propranolol in combination with the glycolysis inhibitor 2DG induced a massive accumulation of autophagosome due to autophagy blockade. The propranolol +2DG treatment efficiently prevents prostate cancer cell proliferation, induces cell apoptosis, alters mitochondrial morphology, inhibits mitochondrial bioenergetics and aggravates ER stress in vitro and also suppresses tumor growth in vivo. Our study underlines for the first time the interest to take advantage of the ability of propranolol to inhibit autophagy to design new anti-cancer therapies.
Collapse
|
80
|
Haldar R, Ben-Eliyahu S. Reducing the risk of post-surgical cancer recurrence: a perioperative anti-inflammatory anti-stress approach. Future Oncol 2018; 14:1017-1021. [PMID: 29623735 DOI: 10.2217/fon-2017-0635] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Rita Haldar
- Sagol School of Neuroscience & School of Psychological Science, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience & School of Psychological Science, Tel-Aviv University, 69978 Tel-Aviv, Israel
| |
Collapse
|
81
|
Liao X, Chaudhary P, Qiu G, Che X, Fan L. The role of propranolol as a radiosensitizer in gastric cancer treatment. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:639-645. [PMID: 29636598 PMCID: PMC5880513 DOI: 10.2147/dddt.s160865] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose The National Comprehensive Cancer Network guidelines indicate that radiotherapy in gastric cancer shows limited effectiveness at reducing the growth of gastric cancer. Therefore, enhancing the sensitivity and effect of radiotherapy with propranolol, a β-adrenoceptor antagonist, could reduce tumor growth. The role of propranolol as a radiosensitizer has not been adequately studied; therefore, the purpose of the present study is to evaluate the effect of propranolol as a radiosensitizer against gastric cancer in vivo. Methods Sixty-four male nude mice bearing tumor xenografts were randomly divided into four groups. Cell culture was performed using the human gastric adenocarcinoma cell line SGC-7901. Mice with tumor xenografts were treated with propranolol, isoproterenol, and radiation. The data for tumor weight and volume were obtained for statistical analyses. Furthermore, the expression levels of COX-2, NF-κB, VEGF, and EGFR were examined using immunohistochemical techniques and Western blotting. Results The growth in the volume and weight of the tumor was lower in mouse models treated with propranolol and radiation therapy compared to the other groups. Decreased expression of NF-κB was also observed in treatment groups where both propranolol and radiation were used, leading to the reduction of COX-2, EGFR, and VEGF expression compared to that in the other groups. Conclusion The present study indicated that propranolol potentiates the antitumor effects of radiotherapy in gastric cancer by inhibiting NF-κB expression and its downstream genes: VEGF, EGFR, and COX-2.
Collapse
Affiliation(s)
- Xinhua Liao
- General Surgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Prakash Chaudhary
- General Surgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guanglin Qiu
- General Surgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiangming Che
- General Surgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lin Fan
- General Surgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
82
|
Jean Wrobel L, Bod L, Lengagne R, Kato M, Prévost-Blondel A, Le Gal FA. Propranolol induces a favourable shift of anti-tumor immunity in a murine spontaneous model of melanoma. Oncotarget 2018; 7:77825-77837. [PMID: 27788481 PMCID: PMC5363624 DOI: 10.18632/oncotarget.12833] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/12/2016] [Indexed: 12/16/2022] Open
Abstract
In a previous study on a xenograft model of melanoma, we showed that the beta-adrenergic receptor antagonist propranolol inhibits melanoma development by modulating angiogenesis, proliferation and cell survival. Stress hormones can influence tumor development in different ways and norepinephrine was shown to downregulate antitumor immune responses by favoring the accumulation of immunosuppressive cells, impairing the function of lymphocytes. We assessed the effect of propranolol on antitumor immune response in the MT/Ret mouse model of melanoma. Propranolol treatment delayed primary tumor growth and metastases development in MT/Ret mice. Consistent with our previous observations in human melanoma xenografts, propranolol induces a decrease in cell proliferation and vessel density in the primary tumors and in metastases. In this immunocompetent model, propranolol significantly reduced the infiltration of myeloid cells, particularly neutrophils, in the primary tumor. Inversely, cytotoxic tumor infiltrating lymphocytes were more frequent in the tumor stroma of treated mice. In a consistent manner, we observed the same shift in the proportions of infiltrating leukocytes in the metastases of treated mice. Our results suggest that propranolol, by decreasing the infiltration of immunosuppressive myeloid cells in the tumor microenvironment, restores a better control of the tumor by cytotoxic cells.
Collapse
Affiliation(s)
- Ludovic Jean Wrobel
- Hôpitaux Universitaires de Genève, Service de Dermatologie, Genève, Switzerland
| | - Lloyd Bod
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Renée Lengagne
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Armelle Prévost-Blondel
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | | |
Collapse
|
83
|
Montoya A, Amaya CN, Belmont A, Diab N, Trevino R, Villanueva G, Rains S, Sanchez LA, Badri N, Otoukesh S, Khammanivong A, Liss D, Baca ST, Aguilera RJ, Dickerson EB, Torabi A, Dwivedi AK, Abbas A, Chambers K, Bryan BA, Nahleh Z. Use of non-selective β-blockers is associated with decreased tumor proliferative indices in early stage breast cancer. Oncotarget 2018; 8:6446-6460. [PMID: 28031536 PMCID: PMC5351644 DOI: 10.18632/oncotarget.14119] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/13/2016] [Indexed: 12/13/2022] Open
Abstract
Previous studies suggest beta-adrenergic receptor (β-AR) antagonists (β-blockers) decrease breast cancer progression, tumor metastasis, and patient mortality; however the mechanism for this is unknown. Immunohistochemical analysis of normal and malignant breast tissue revealed overexpression of β1-AR and β3-AR in breast cancer. A retrospective cross-sectional study of 404 breast cancer patients was performed to determine the effect of β-blocker usage on tumor proliferation. Our analysis revealed that non-selective β-blockers, but not selective β-blockers, reduced tumor proliferation by 66% (p < 0.0001) in early stage breast cancer compared to non-users. We tested the efficacy of propranolol on an early stage breast cancer patient, and quantified the tumor proliferative index before and after treatment, revealing a propranolol-mediated 23% reduction (p = 0.02) in Ki67 positive tumor cells over a three-week period. The anti-proliferative effects of β-blockers were measured in a panel of breast cancer lines, demonstrating that mammary epithelial cells were resistant to propranolol, and that most breast cancer cell lines displayed dose dependent viability decreases following treatment. Selective β-blockers alone or in combination were not as effective as propranolol at reducing breast cancer cell proliferation. Molecular analysis revealed that propranolol treatment of the SK-BR-3 breast cancer line, which showed high sensitivity to beta blockade, led to a reduction in Ki67 protein expression, decreased phosphorylation of the mitogenic signaling regulators p44/42 MAPK, p38 MAPK, JNK, and CREB, increased phosphorylation of the cell survival/apoptosis regulators AKT, p53, and GSK3β. In conclusion, use of non-selective β-blockers in patients with early stage breast cancer may lead to decreased tumor proliferation.
Collapse
Affiliation(s)
- Alexa Montoya
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Department of Biology, University of Texas, El Paso, Texas, USA
| | - Clarissa N Amaya
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Andres Belmont
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Nabih Diab
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Richard Trevino
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Geri Villanueva
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Steven Rains
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Luis A Sanchez
- Department of Hematology/Oncology, Loma Linda University Health Sciences Center, Loma Linda, California, USA
| | - Nabeel Badri
- Department of Hematology/Oncology, Loma Linda University Health Sciences Center, Loma Linda, California, USA
| | - Salman Otoukesh
- Department of Hematology/Oncology, Loma Linda University Health Sciences Center, Loma Linda, California, USA
| | - Ali Khammanivong
- Department of Veterinary Clinical Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Danielle Liss
- Department of Hematology/Oncology, Loma Linda University Health Sciences Center, Loma Linda, California, USA
| | - Sarah T Baca
- Border Biomedical Research Center, University of Texas, El Paso, Texas, USA
| | - Renato J Aguilera
- Border Biomedical Research Center, University of Texas, El Paso, Texas, USA
| | - Erin B Dickerson
- Department of Veterinary Clinical Sciences, University of Minnesota, Saint Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alireza Torabi
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Department of Pathology, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Alok K Dwivedi
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Division of Biostatistics and Epidemiology, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Aamer Abbas
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Department of Hematology/Oncology, Loma Linda University Health Sciences Center, Loma Linda, California, USA
| | - Karinn Chambers
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Department of Surgery, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Brad A Bryan
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Zeina Nahleh
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA.,Department of Hematology/Oncology, Loma Linda University Health Sciences Center, Loma Linda, California, USA
| |
Collapse
|
84
|
Qiao G, Chen M, Bucsek MJ, Repasky EA, Hylander BL. Adrenergic Signaling: A Targetable Checkpoint Limiting Development of the Antitumor Immune Response. Front Immunol 2018; 9:164. [PMID: 29479349 PMCID: PMC5812031 DOI: 10.3389/fimmu.2018.00164] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/18/2018] [Indexed: 12/15/2022] Open
Abstract
An immune response must be tightly controlled so that it will be commensurate with the level of response needed to protect the organism without damaging normal tissue. The roles of cytokines and chemokines in orchestrating these processes are well known, but although stress has long been thought to also affect immune responses, the underlying mechanisms were not as well understood. Recently, the role of nerves and, specifically, the sympathetic nervous system, in regulating immune responses is being revealed. Generally, an acute stress response is beneficial but chronic stress is detrimental because it suppresses the activities of effector immune cells while increasing the activities of immunosuppressive cells. In this review, we first discuss the underlying biology of adrenergic signaling in cells of both the innate and adaptive immune system. We then focus on the effects of chronic adrenergic stress in promoting tumor growth, giving examples of effects on tumor cells and immune cells, explaining the methods commonly used to induce stress in preclinical mouse models. We highlight how this relates to our observations that mandated housing conditions impose baseline chronic stress on mouse models, which is sufficient to cause chronic immunosuppression. This problem is not commonly recognized, but it has been shown to impact conclusions of several studies of mouse physiology and mouse models of disease. Moreover, the fact that preclinical mouse models are chronically immunosuppressed has critical ramifications for analysis of any experiments with an immune component. Our group has found that reducing adrenergic stress by housing mice at thermoneutrality or treating mice housed at cooler temperatures with β-blockers reverses immunosuppression and significantly improves responses to checkpoint inhibitor immunotherapy. These observations are clinically relevant because there are numerous retrospective epidemiological studies concluding that cancer patients who were taking β-blockers have better outcomes. Clinical trials testing whether β-blockers can be repurposed to improve the efficacy of traditional and immunotherapies in patients are on the horizon.
Collapse
Affiliation(s)
- Guanxi Qiao
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Minhui Chen
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mark J. Bucsek
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Elizabeth A. Repasky
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bonnie L. Hylander
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
85
|
Gandini S, Palli D, Spadola G, Bendinelli B, Cocorocchio E, Stanganelli I, Miligi L, Masala G, Caini S. Anti-hypertensive drugs and skin cancer risk: a review of the literature and meta-analysis. Crit Rev Oncol Hematol 2018; 122:1-9. [PMID: 29458778 DOI: 10.1016/j.critrevonc.2017.12.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/04/2017] [Accepted: 12/06/2017] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Several anti-hypertensive drugs have photosensitizing properties, however it remains unclear whether long-term users of these drugs are also at increased risk of skin malignancies. We conducted a literature review and meta-analysis on the association between use of anti-hypertensive drugs and the risk of cutaneous melanoma and non-melanoma skin cancer (NMSC). METHODS We searched PubMed, EMBASE, Google Scholar and the Cochrane Library, and included observational and experimental epidemiological studies published until February 28th, 2017. We calculated summary relative risk (SRR) and 95% confidence intervals (95% CI) through random effect models to estimate the risk of skin malignancies among users of the following classes of anti-hypertensive drugs: thiazide diuretics, angiotensin converting enzyme inhibitors (ACEi), angiotensin receptor blockers (ARB), calcium channel blockers (CCB) and β-blockers. We conducted sub-group and sensitivity analysis to explore causes of between-studies heterogeneity, and assessed publication bias using a funnel-plot based approach. RESULTS Nineteen independent studies were included in the meta-analysis. CCB users were at increased skin cancer risk (SRR 1.14, 95% CI 1.07-1.21), and β-blockers users were at increased risk of developing cutaneous melanoma (SRR 1.21, 95% CI 1.05-1.40), with acceptable between-studies heterogeneity (I2 < 50%). There was no association between thiazide diuretics, ACEi or ARB use and skin cancer risk. We found no evidence of publication bias affecting the results. CONCLUSION Family doctors and clinicians should inform their patients about the increased risk of skin cancer associated with the use of CCB and β-blockers and instruct them to perform periodic skin self-examination. Further studies are warranted to elucidate the observed associations.
Collapse
Affiliation(s)
- Sara Gandini
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - Domenico Palli
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Giuseppe Spadola
- Division of Melanoma and Muscolo-Cutaneous Sarcoma, European Institute of Oncology, Milan, Italy
| | - Benedetta Bendinelli
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Emilia Cocorocchio
- Division of Melanoma and Muscolo-Cutaneous Sarcoma, European Institute of Oncology, Milan, Italy
| | - Ignazio Stanganelli
- Skin Cancer Unit, IRCCS-IRST Scientific Institute of Romagna for the Study and Treatment of Cancer, Meldola, Italy
| | - Lucia Miligi
- Environmental and Occupational Epidemiology Branch, Cancer Risk Factors and Lifestyle Epidemiology Unit, Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Giovanna Masala
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Saverio Caini
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Cancer Research and Prevention Institute (ISPO), Florence, Italy.
| |
Collapse
|
86
|
Wang B, Parobchak N, Martin A, Rosen M, Yu LJ, Nguyen M, Gololobova K, Rosen T. Screening a small molecule library to identify inhibitors of NF-κB inducing kinase and pro-labor genes in human placenta. Sci Rep 2018; 8:1657. [PMID: 29374256 PMCID: PMC5785954 DOI: 10.1038/s41598-018-20147-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/15/2018] [Indexed: 01/17/2023] Open
Abstract
The non-canonical NF-κB signaling (RelB/p52) pathway drives pro-labor genes in the human placenta, including corticotropin-releasing hormone (CRH) and cyclooxygenase-2 (COX-2), making this a potential therapeutic target to delay onset of labor. Here we sought to identify small molecule compounds from a pre-existing chemical library of orally active drugs that can inhibit this NF-κB signaling, and in turn, human placental CRH and COX-2 production. We used a cell-based assay coupled with a dual-luciferase reporter system to perform an in vitro screening of a small molecule library of 1,120 compounds for inhibition of the non-canonical NF-κB pathway. Cell toxicity studies and drug efflux transport MRP1 assays were used to further characterize the lead compounds. We have found that 14 drugs have selective inhibitory activity against lymphotoxin beta complex-induced activation of RelB/p52 in HEK293T cells, several of which also inhibited expression of CRH and COX-2 in human term trophoblast. We identified sulfapyridine and propranolol with activity against CRH and COX-2 that deserve further study. These drugs could serve as the basis for development of orally active drugs to affect length of gestation, first in an animal model, and then in clinical trials to prevent preterm birth during human pregnancy.
Collapse
Affiliation(s)
- Bingbing Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA.
| | - Nataliya Parobchak
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Adriana Martin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Max Rosen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Lumeng Jenny Yu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Mary Nguyen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Kseniya Gololobova
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Todd Rosen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA.
| |
Collapse
|
87
|
Wang F, Liu H, Wang F, Xu R, Wang P, Tang F, Zhang X, Zhu Z, Lv H, Han T. Propranolol suppresses the proliferation and induces the apoptosis of liver cancer cells. Mol Med Rep 2018; 17:5213-5221. [PMID: 29393410 PMCID: PMC5865987 DOI: 10.3892/mmr.2018.8476] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/13/2017] [Indexed: 12/26/2022] Open
Abstract
An increasing amount of evidence indicates that the inhibition of β adrenergic signaling can result in the inhibition of tumor growth. However, the role of propranolol in liver cancer and the underlying mechanism remain to be elucidated. The present study aimed to investigate the role of propranolol in liver cancer cell lines and provide evidence for further clinical study. Propranolol was added at different concentrations to HepG2 and HepG2.2.15 liver cancer cells and HL-7702 normal human liver cells. The proliferation of the cell lines was monitored by live-cell imaging at a range of time intervals. Immunofluorescence using DAPI and Hoechst 33342/propidium iodide (PI) staining, Annexin V-FITC/PI double-staining flow cytometry, western blotting and reverse transcription-quantitative polymerase chain reaction were used to investigate the effect of propranolol on liver cancer cell apoptosis. The proliferation of HepG2 and HepG2.2.15 cells was inhibited by 40 and 80 µmol/l propranolol. However, the proliferation of HL-7702 cells was not affected by <160 µmol/l propranolol. Propranolol treatment decreased the expression of adrenergic receptor β-2 to a greater extent than adrenergic receptor β-1, and induced apoptosis in the liver cancer cells. The apoptotic rates of HepG2 and HepG2.2.15 cells increased following treatment with propranolol, while the apoptotic rate of HL-7702 cells was not affected. Propranolol promoted poly (ADP-ribose) polymerase cleavage and decreased the expression of full-length caspase-3 in liver cancer cell lines; it induced S-phase arrest in HepG2 and HepG2.2.15 cell lines, while HL-7702 cells were arrested at the G0/G1 phase of the cell cycle. Thus, it was demonstrated that propranolol inhibited proliferation, promoted apoptosis and induced S-phase arrest in HepG2 and HepG2.2.15 cells.
Collapse
Affiliation(s)
- Fang Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, P.R. China
| | - Hui Liu
- Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, P.R. China
| | - Fengmei Wang
- Department of Gastroenterology and Hepatology of Tianjin Third Central Hospital, Tianjin 300170, P.R. China
| | - Ruicheng Xu
- Tianjin Key Laboratory for Biomarkers of Occupational and Environmental Hazard, Tianjin 300170, P.R. China
| | - Peng Wang
- Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, P.R. China
| | - Fei Tang
- Department of Gastroenterology and Hepatology of Tianjin Third Central Hospital, Tianjin 300170, P.R. China
| | - Xu Zhang
- Department of Gastroenterology and Hepatology of Tianjin Third Central Hospital, Tianjin 300170, P.R. China
| | - Zhengyan Zhu
- Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, P.R. China
| | - Hongmin Lv
- Department of Gastroenterology and Hepatology of Tianjin Third Central Hospital, Tianjin 300170, P.R. China
| | - Tao Han
- Department of Gastroenterology and Hepatology of Tianjin Third Central Hospital, Tianjin 300170, P.R. China
| |
Collapse
|
88
|
Wick W, Kessler T. Drug Repositioning Meets Precision in Glioblastoma. Clin Cancer Res 2017; 24:256-258. [PMID: 29133572 DOI: 10.1158/1078-0432.ccr-17-2989] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 10/28/2017] [Accepted: 11/07/2017] [Indexed: 11/16/2022]
Abstract
Glioblastoma has a gigantic unmet medical need. Molecular knowledge has evolved substantially, including data on clonal selection with progression. Past trials for all-comers may have produced false negative results. Molecular precision at progression needs workup of new tissue, and revisiting drugs with a focus on brain tumor penetration may yield surprises. Clin Cancer Res; 24(2); 256-8. ©2017 AACRSee related article by Byron et al., p. 295.
Collapse
Affiliation(s)
- Wolfgang Wick
- Neurology Clinic, University of Heidelberg, Heidelberg, Germany. .,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Kessler
- Neurology Clinic, University of Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
89
|
Ni H, Rui Q, Zhu X, Yu Z, Gao R, Liu H. Antihypertensive drug use and breast cancer risk: a meta-analysis of observational studies. Oncotarget 2017; 8:62545-62560. [PMID: 28977968 PMCID: PMC5617528 DOI: 10.18632/oncotarget.19117] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 04/14/2017] [Indexed: 12/14/2022] Open
Abstract
We conducted a meta-analysis of observational studies to examine the hypothesized association between breast cancer and antihypertensive drug (AHT) use. Fixed- or random- effect models were used to calculate pooled risk ratios (RRs) and 95% confidence intervals (CIs) for all AHTs and individual classes (i.e., angiotensin-converting enzyme inhibitors, [ACEi]; angiotensin-receptor blockers, [ARBs]; calcium channel blockers, [CCBs]; beta-blockers, [BBs], and diuretics). Twenty-one studies with 3,116,266 participants were included. Overall, AHT use was not significantly associated with breast cancer risk (RR = 1.02, 95% CI: 0.98-1.06), and no consistent association was found for specific AHT classes with pooled RRs of 1.02 (95% CI: 0.96-1.09) for BBs, 1.07 (95% CI: 0.99-1.16) for CCBs, 0.99 (95% CI: 0.93-1.05) for ACEi/ARBs, and 1.05 (95% CI: 0.99-1.12) for diuretics. When stratified by duration of use, there was a significantly reduced breast cancer risk for ACEi/ARB use ≥10 years (RR = 0.80, 95% CI: 0.67-0.95). Although there was no significant association between AHT use and breast cancer risk, there was a possible beneficial effect was found for long-term ACEi/ARB. Large, randomized controlled trials with long-term follow-up are needed to further test the effect of these medications on breast cancer risk.
Collapse
Affiliation(s)
- Haibo Ni
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang City, Suzhou, Jiangsu, China
| | - Qin Rui
- Department of Laboratory, The First People's Hospital of Zhangjiagang City, Suzhou, Jiangsu, China
| | - Xiaojue Zhu
- Department of Laboratory, The First People's Hospital of Zhangjiagang City, Suzhou, Jiangsu, China
| | - Zhenquan Yu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rong Gao
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang City, Suzhou, Jiangsu, China
| | - Huixiang Liu
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang City, Suzhou, Jiangsu, China
| |
Collapse
|
90
|
Harris DA, Park JM, Lee KS, Xu C, Stella N, Hague C. Label-Free Dynamic Mass Redistribution Reveals Low-Density, Prosurvival α1B-Adrenergic Receptors in Human SW480 Colon Carcinoma Cells. J Pharmacol Exp Ther 2017; 361:219-228. [PMID: 28196836 PMCID: PMC5399639 DOI: 10.1124/jpet.116.237255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 02/10/2017] [Indexed: 12/11/2022] Open
Abstract
Small molecules that target the adrenergic family of G protein-coupled receptors (GPCRs) show promising therapeutic efficacy for the treatment of various cancers. In this study, we report that human colon cancer cell line SW480 expresses low-density functional α1B-adrenergic receptors (ARs) as revealed by label-free dynamic mass redistribution (DMR) signaling technology and confirmed by quantitative reverse-transcriptase polymerase chain reaction analysis. Remarkably, although endogenous α1B-ARs are not detectable via either [3H]-prazosin-binding analysis or phosphoinositol hydrolysis assays, their activation leads to robust DMR and enhanced cell viability. We provide pharmacological evidence that stimulation of α1B-ARs enhances SW480 cell viability without affecting proliferation, whereas stimulating β-ARs diminishes both viability and proliferation of SW480 cells. Our study illustrates the power of label-free DMR technology for identifying and characterizing low-density GPCRs in cells and suggests that drugs targeting both α1B- and β-ARs may represent valuable small-molecule therapeutics for the treatment of colon cancer.
Collapse
Affiliation(s)
- Dorathy-Ann Harris
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Ji-Min Park
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Kyung-Soon Lee
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Cong Xu
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Nephi Stella
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Chris Hague
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
91
|
Iannaccone M, D´Ascenzo F, De Filippo O, Gagliardi M, Southern DA, Raposeiras-Roubín S, Abu-Assi E, Henriques JPS, Saucedo J, González-Juanatey JR, Wilton SB, Kikkert WJ, Nuñez-Gil I, Ariza-Sole A, Song X, Alexopoulos D, Liebetrau C, Kawaji T, Huczek Z, Nie SP, Fujii T, Correia L, Kawashiri MA, García-Acuña JM, Alfonso E, Terol B, Garay A, Zhang D, Chen Y, Xanthopoulou I, Osman N, Möllmann H, Shiomi H, Kowara M, Filipiak K, Wang X, Yan Y, Fan JY, Ikari Y, Nakahashi T, Sakata K, Yamagishi M, Moretti C, Gaita F, Kalpak O, Kedev S. Optimal Medical Therapy in Patients with Malignancy Undergoing Percutaneous Coronary Intervention for Acute Coronary Syndrome: a BleeMACS Sub-Study. Am J Cardiovasc Drugs 2017; 17:61-71. [PMID: 27738920 DOI: 10.1007/s40256-016-0196-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Our objective was to define the most appropriate treatment for acute coronary syndrome (ACS) in patients with malignancy. METHODS AND RESULTS The BleeMACS project is a worldwide multicenter observational prospective registry in 16 hospitals enrolling patients with ACS undergoing percutaneous coronary intervention. Primary endpoints were death, re-infarction, and major adverse cardiac events (MACE; composite of death and re-infarction) after 1 year of follow-up. The secondary endpoint was bleeding events during follow-up. We performed sub-study analyses according to whether β-blockers (BBs), angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs), statins, or proton pump inhibitors (PPIs) were prescribed at discharge. We also calculated the propensity score for optimal medical therapy (OMT; combination of BB, ACEI/ARB, and statins). The study included 926 patients. According to the multivariate analysis, ACEIs/ARBs (hazard ratio [HR] 0.58, 95 % confidence interval [CI] 0.36-1.94; p = 0.03) and statins (HR 0.37, 95 % CI 0.23-0.61; p < 0.01) reduced the risk of MACE, while the effects of BBs (HR 0.85, 95 % CI 0.55-1.32; p = 0.48) and PPIs (HR 1.33, 95 % CI 0.83-2.12; p = 0.23) were not significant. OMT was prescribed at discharge in 300 (32.4 %) patients; after propensity score analysis, OMT showed a significant reduction in death (3 % vs. 12.5 %, HR 0.21, 95 % CI 0.1-0.4; log-rank p < 0.001) and MACE (6.7 vs. 15.2 %, log-rank p = 0.01). CONCLUSION In patients with ACS and malignancy, OMT reduces the risk of adverse events at 1 year; in particular, ACEIs/ARBs and statins were the most protective drugs. (Clinical trials identifier: NCT02466854).
Collapse
|
92
|
Postdiagnostic use of β-blockers and other antihypertensive drugs and the risk of recurrence and mortality in head and neck cancer patients: an observational study of 10,414 person-years of follow-up. Clin Transl Oncol 2017; 19:826-833. [PMID: 28093700 DOI: 10.1007/s12094-016-1608-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Growing evidence indicates that norepinephrine promotes cancer growth and metastasis whereas β-blockers decrease these risks. This study aimed to examine the clinical impact of β-blockers and other hypertensive drugs on disease recurrence and survival in patients with head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS This study analyzed a cohort of 1274 consecutive patients who received definitive treatments for previously untreated HNSCC at our tertiary referral center between January 2001 and December 2012. Antihypertensive use was considered positive if patients were on medication from HNSCC diagnosis to at least 1 year after treatment initiation. Cox proportional hazard models were utilized to determine associations between antihypertensive drugs and recurrence, survival, and second primary cancer (SPC) occurrence. RESULTS Hypertension itself was not a significant variable of recurrence and survival and no antihypertensive drug use affected SPC occurrence (all P > 0.1). After controlling for clinical factors, calcium-channel blocker use remained an independent variable for index cancer recurrence, and β-blocker use was significantly associated with poor cancer-specific mortality, competing mortality, and all-cause mortality (all P < 0.05). β-blocker use significantly affected competing and all-cause mortalities in normotensive patients, and calcium-channel blocker use affected index cancer recurrence in normotensive patients (all P < 0.05). CONCLUSIONS Our data show that β-blocker use is associated with decreased survival and calcium-channel blockers is associated with increased cancer recurrence in patients of HNSCC.
Collapse
|
93
|
Pantziarka P, Bouche G, Sukhatme V, Meheus L, Rooman I, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-Propranolol as an anti-cancer agent. Ecancermedicalscience 2016; 10:680. [PMID: 27899953 PMCID: PMC5102691 DOI: 10.3332/ecancer.2016.680] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Indexed: 12/23/2022] Open
Abstract
Propranolol (PRO) is a well-known and widely used non-selective beta-adrenergic receptor antagonist (beta-blocker), with a range of actions which are of interest in an oncological context. PRO displays effects on cellular proliferation and invasion, on the immune system, on the angiogenic cascade, and on tumour cell sensitivity to existing treatments. Both pre-clinical and clinical evidence of these effects, in multiple cancer types, is assessed and summarised and relevant mechanisms of action outlined. In particular there is evidence that PRO is effective at multiple points in the metastatic cascade, particularly in the context of the post-surgical wound response. Based on this evidence the case is made for further clinical investigation of the anticancer effects of PRO, particularly in combination with other agents. A number of trials are on-going, in different treatment settings for various cancers.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium; The George Pantziarka TP53 Trust, London, UK
| | | | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | - Ilse Rooman
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium; Oncology Research Centre, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc, Newton MA 02459, USA; Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
94
|
Abstract
Stress as a modern civilization factor significantly affects our lives. While acute stress might have a positive effect on the organism, chronic stress is usually detrimental and might lead to serious health complications. It is known that stress induced by the physical environment (temperature-induced cold stress) can significantly impair the efficacy of cytotoxic chemotherapies and the anti-tumor immune response. On the other hand, epidemiological evidence has shown that patients taking drugs known as β-adrenergic antagonists ("β-blockers"), which are commonly prescribed to treat arrhythmia, hypertension, and anxiety, have significantly lower rates of several cancers. In this review, we summarize the current knowledge about catecholamines as important stress hormones in tumorigenesis and discuss the use of β-blockers as the potential therapeutic agents.
Collapse
Affiliation(s)
- O Krizanova
- a Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences , Bratislava , Slovakia
- b Department of Physiology, Faculty of Medicine , Masaryk University , Brno , Czech Republic
| | - P Babula
- b Department of Physiology, Faculty of Medicine , Masaryk University , Brno , Czech Republic
| | - K Pacak
- c Development, Endocrinology, and Tumor Genetics Affinity Group, Section on Medical Neuroendocrinology , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
95
|
Zhang L, Nie Y, Zheng Y, Ke L, Tong Z, Li W, Li J. Esmolol attenuates lung injury and inflammation in severe acute pancreatitis rats. Pancreatology 2016; 16:726-32. [PMID: 27269252 DOI: 10.1016/j.pan.2016.05.395] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 05/18/2016] [Accepted: 05/29/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recent studies suggest that beta-adrenergic blockers attenuate systemic inflammation and improve survival in sepsis. We investigated whether esmolol can reduce lung injury and modulate inflammatory response in a rat model of severe acute pancreatitis (SAP). METHODS A taurocholate-induced SAP was used, with or without continuously intravenous pumping of esmolol (15 mg/kg/h). Heart rate and arterial pressure were monitored. Nine hrs after esmolol administration, blood was drawn for blood gas analyses and cytokine (interleukin(IL)-6, tumor necrosis factor (TNF)-α) detections, lungs and pancreata were isolated for measurements of myeloperoxidase (MPO) activity and histological damage. In an additional 20 animals, rats were randomized into SAP or SAP + esmolol groups to assess effects of esmolol on survival time. RESULTS Treatment with esmolol was associated with improved survival time (11.1 ± 1.6 h vs. 9.2 ± 2.0 h, p = 0.044) and less severe disease, as assessed by lung and pancreas histology. Blood gas analyses were ameliorated in esmolol group. Arterial PO2 increased (109.7 ± 12.4 mmHg vs 93.9 ± 4.1 mmHg, p = 0.008) while lactate levels (2.1 ± 0.5 vs 3.1 ± 0.7 mmol/L, p = 0.001) decreased in SAP + esmolol group as compared with SAP group. Esmolol treatment also abated the increase in bronchoalveolar lavage fluid protein and proinflammatory cytokines. Furthermore, esmolol reduced SAP-induced plasma amylase activity (p = 0.02), blunted the expression of TNF-α (p = 0.003) and IL-6 (p < 0.001), and decreased pancreas/lung MPO activities. CONCLUSIONS Continuous infusion of esmolol, a selective beta-1 adrenergic blocker, improves outcome, reduces inflammatory responses and also offers lung and pancreas protective effects in SAP rats. This may offer novel therapeutic strategies in treating patients suffering from SAP.
Collapse
Affiliation(s)
- Luyao Zhang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China; School of Basic Medical Science, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Yao Nie
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yishan Zheng
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lu Ke
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhihui Tong
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiqin Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Jieshou Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|