1
|
Chen CH, Reva B, Katabi N, Wizel A, Xu H, Ho AL, Morris LG, Bakst RL, Parikh AS, Drier Y, Deborde S, Wong RJ. Sympathetic axonogenesis promotes adenoid cystic carcinoma progression. J Exp Med 2025; 222:e20242250. [PMID: 40272482 PMCID: PMC12020745 DOI: 10.1084/jem.20242250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/07/2025] [Accepted: 03/12/2025] [Indexed: 04/25/2025] Open
Abstract
Nerves are integral to the adenoid cystic carcinoma (ACC) microenvironment. The strong association of ACC with perineural invasion (PNI) is considered a hallmark of this disease. In human salivary ACC, we identify intratumoral, small-caliber, disorganized sympathetic nerves not observed in other salivary neoplasms. Norepinephrine or sympathetic ganglia explants enhance ACC proliferation in vitro. Two novel orthotopic ACC patient-derived xenograft (PDX) models recapitulate ACC morphology and demonstrate sympathetic innervation. Pharmacologic or surgical blockade of sympathetic nerves decreases ACC PDX growth. Bulk RNA sequencing of salivary ACC reveals correlations between noradrenergic nerve development signatures and worse patient survival. Metastatic ACC foci exhibit lower nerve signature gene expression levels than primary ACC. Sympathetic innervation in ACC is distinct from PNI and reflects tumor axonogenesis driven by noradrenergic neural development programs. These programs support ACC progression, are associated with poor prognosis, and may be inhibited as a therapeutic strategy.
Collapse
Affiliation(s)
- Chun-Hao Chen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Boris Reva
- Department of Genetics and Genomic Sciences, Mount Sinai Medical Center, New York, NY, USA
| | - Nora Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Avishai Wizel
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hongbo Xu
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Alan L. Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc G.T. Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard L. Bakst
- Department of Radiation Oncology, Mount Sinai Medical Center, New York, NY, USA
| | - Anuraag S. Parikh
- Department of Otolaryngology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Yotam Drier
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sylvie Deborde
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard J. Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
2
|
Palanisamy B, Mandal AKA. Unlocking the potential: Receptor-mediated targeted drug delivery in cancer therapy. Pathol Res Pract 2025; 270:155955. [PMID: 40209568 DOI: 10.1016/j.prp.2025.155955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/29/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Receptor-mediated targeted drug delivery has emerged as a pivotal strategy in cancer therapy, offering precision and specificity in combating malignant diseases while minimizing systemic toxicity. This review explores the multifaceted role of receptors in cancer biology, emphasizing their contributions to cancer progression, metastasis, and their potential as therapeutic targets. Ligand-based targeting approaches highlight the utility of small molecules, peptides, and antibodies, as well as the development of novel targeting ligands. A critical focus is placed on engineering receptor-targeted nanoparticles and advanced drug delivery systems. Innovations in dual-targeting strategies and the targeted delivery to the tumour microenvironment (TME) and metastatic niches are discussed, underscoring their potential to enhance therapeutic efficacy. Additionally, receptor-targeted imaging is reviewed for its dual role in diagnosis and real-time treatment monitoring. To address the challenges of side effects and off-target toxicity, strategies that minimize these risks while targeting overexpressed receptors in solid tumours are explored. Finally, the review outlines future directions in receptor-targeted cancer therapy, emphasizing the need for interdisciplinary research to refine these strategies further. This comprehensive analysis aims to provide a roadmap for advancing receptor-based therapeutic approaches, ultimately improving outcomes for cancer patients.
Collapse
Affiliation(s)
- Balaji Palanisamy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| | - Abul Kalam Azad Mandal
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
3
|
Chen Z, Zhou Y, Xue C, Zeng L, Deng S, Xu Z, Li M, Zhao H, He X, Liu S, Liu J, Liu S, Zhao S, Zhang S, Peng X, Wu X, Bai R, Zhuang L, Wu S, Zhang J, Lin D, Huang X, Zheng J. Psychological stress-induced ALKBH5 deficiency promotes tumour innervation and pancreatic cancer via extracellular vesicle transfer of RNA. Nat Cell Biol 2025:10.1038/s41556-025-01667-0. [PMID: 40419796 DOI: 10.1038/s41556-025-01667-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 04/02/2025] [Indexed: 05/28/2025]
Abstract
The pathological role and mechanism of psychological stress in cancer progression are little known. Here we show in a mouse model that psychological stress drives pancreatic ductal adenocarcinoma (PDAC) progression by stimulating tumour nerve innervation. We demonstrate that nociception and other stressors activate sympathetic nerves to release noradrenaline, downregulating RNA demethylase alkB homologue 5 (Alkbh5) in tumour cells. Alkbh5 deficiency in these cancer cells causes aberrant N6-methyladenosine (m6A) modification of RNAs, which are packed into extracellular vesicles and delivered to nerves in the tumour microenvironment, enhancing hyperinnervation and PDAC progression. ALKBH5 levels are inversely correlated with tumour innervation and survival time in patients with PDAC. Animal experiments identify a natural flavonoid, fisetin, that prevents neurons from taking in extracellular vesicles containing m6A-modified RNAs, thus suppressing the excessive innervation and progression of PDAC tumours. Our study sheds light on a molecular mechanism by which crosstalk between the neuroendocrine system and cancer cells links psychological stress and cancer progression and raises a potential strategy for PDAC therapy.
Collapse
Affiliation(s)
- Ziming Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yifan Zhou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Chunling Xue
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Lingxing Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shuang Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Zilan Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Mei Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongzhe Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xiaowei He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaoqiu Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ji Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shuang Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Sihan Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaoping Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xinyi Peng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xiaoyu Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ruihong Bai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Lisha Zhuang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaojia Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jialiang Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Dongxin Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Xudong Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
| | - Jian Zheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Caller T, Moore KJ, Lehmann LH, Wu SM, Leor J. Insights Into Heart-Tumor Interactions in Heart Failure. Circ Res 2025; 136:1262-1285. [PMID: 40403117 DOI: 10.1161/circresaha.124.325490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/10/2025] [Accepted: 02/27/2025] [Indexed: 05/24/2025]
Abstract
Heart failure (HF) often coexists with cancer. Beyond the known cardiotoxicity of some cancer treatments, HF itself has been associated with increased cancer incidence. The 2 conditions share common risk factors, mechanisms, and interactions that can worsen patient outcomes. The bidirectional relationship between HF and cancer presents a complex interplay of factors that are not fully understood. Recent preclinical evidence suggests that HF may promote tumor growth via the release of protumorigenic factors from the injured heart, revealing HF as a potentially protumorigenic condition. Our review discusses the biological crosstalk between HF and cancer, emphasizing the impact of HF on tumor growth, with inflammation, and modulating the immune system as central mechanisms. We further explore the clinical implications of this connection and propose future research directions. Understanding the mechanistic overlap and interactions between HF and cancer could lead to new biomarkers and therapies, addressing the growing prevalence of both conditions and enhancing approaches to diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Tal Caller
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| | - Kathryn J Moore
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine (K.J.M.)
| | - Lorenz H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Germany (L.H.L.)
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg/Mannheim, Germany (L.H.L.)
- German Cancer Research Center (DKFZ), Heidelberg, Germany (L.H.L.)
| | - Sean M Wu
- Stanford Cardiovascular Institute (S.M.W.), Stanford University School of Medicine, CA
- Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA
| | - Jonathan Leor
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| |
Collapse
|
5
|
Repasky E, Mohammadpour H. Targeting nerve growth factor: an Achilles' heel for tumors? J Immunother Cancer 2025; 13:e011609. [PMID: 40316307 PMCID: PMC12049907 DOI: 10.1136/jitc-2025-011609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/01/2025] [Indexed: 05/04/2025] Open
Abstract
A tumor's ability to attract innervation is a critical factor in tumor progression and immune escape, with the sympathetic nervous system playing a major role. Catecholamines released by sympathetic nerves activate adrenergic receptors on tumor cells, enhancing growth and resistance to therapies, while activation of adrenergic receptors on immune cells triggers immunosuppressive activity in the tumor microenvironment. Nerve growth factor (NGF) produced by tumor cells is a key driver of tumor innervation, making it a promising target for novel therapeutic strategies. In this commentary, we highlight a recent study by Yang et al, which examines NGF single-chain variable fragment (scFv)-secreting chimeric antigen receptor(CAR) T cells and the impact of NGF neutralization by CAR T cells on CAR T-cell function and the remodeling of the tumor microenvironment. This work shows that we may be able to exploit a tumor-derived survival factor as a vulnerability and a means to enhance antitumor immune activity.
Collapse
Affiliation(s)
- Elizabeth Repasky
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Hemn Mohammadpour
- Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
6
|
Krishnamoorthy S, Vasudevan S, Muruganantham B, Muthusami S. Epinephrine augments the phosphorylation of EGFR and promote the DNA synthesis and migration of cervical cancer cells. 3 Biotech 2025; 15:125. [PMID: 40242059 PMCID: PMC11996747 DOI: 10.1007/s13205-025-04285-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Though a correlation exists between carcinogenesis and epinephrine signaling, the ability of epinephrine in regulating epidermal growth factor (EGF) actions remains largely unknown and necessitates investigation in cervical cancer (CC) cells. The present study aims to understand the role of epinephrine, a stress-induced cytokine on EGF actions in human papilloma virus (HPV)-positive SiHa, ME180 and HPV-negative C33A cells. De-identified database and molecular docking were used to identify the relationship between beta-adrenergic receptor 2 (ADRB2) and EGF receptor (EGFR). Cell viability, mitochondrial labeling, reactive oxygen species (ROS) production, evaluation of ADRB2 and superoxide dismutase-2 (SOD-2) mRNA abundance, SOD-1/2 enzymatic activities, migration assay, gelatin zymography and protein expression analysis of epithelial-mesenchymal transition (EMT) markers were performed to validate the regulatory role of epinephrine and EGF. A significant up-regulation of ADRB2 in HPV-16-positive individuals and binding between epinephrine and EGFR is noted computationally. A reduced survival in high ADRB2 along with the significant reduction in CC survival in Asian population is also observed. Epinephrine augmented the phosphorylation of EGFR and EGF-induced cell viability, ROS production and DNA synthesis. A positive correlation between SOD-2 and ADRB2 was corroborated with the increased ADRB2 and SOD-2 mRNA transcripts. An increase in MMP-2 activity and EMT markers by EGF and epinephrine potentiated the CC cells toward enhanced migration. This study also opens up several new avenues and warrants substantial in vivo studies to support this contention for the inclusion of beta-blockers as adjuvant for EGFR-driven cancers.
Collapse
Affiliation(s)
- Sneha Krishnamoorthy
- Department of Biochemistry, Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641 021 India
| | - Saraswathi Vasudevan
- Department of Biochemistry, Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641 021 India
| | - Bharathi Muruganantham
- Department of Biochemistry, Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641 021 India
- Centre for Bioinformatics, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641 021 India
| | - Sridhar Muthusami
- Department of Biochemistry, Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641 021 India
- Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641 021 India
| |
Collapse
|
7
|
Alzahrani SM, Al Doghaither HA, Alkhatabi HA, Basabrain MA, Pushparaj PN. Propranolol and Capecitabine Synergy on Inducing Ferroptosis in Human Colorectal Cancer Cells: Potential Implications in Cancer Therapy. Cancers (Basel) 2025; 17:1470. [PMID: 40361395 PMCID: PMC12071015 DOI: 10.3390/cancers17091470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal cancer (CRC) is a significant global health issue with rising incidence and mortality rates. In oncology, drug repurposing has emerged as a promising therapeutic strategy in conjunction with conventional treatments. This study aimed to evaluate the potential of repurposing propranolol (PRO), a beta blocker, for the treatment of CRC cell lines (HCT-116 and HT-29), both as a monotherapy and in combination with capecitabine (CAP). METHODS Effects of mono- and combination therapies on viability, combination index, morphology, and cell death induction of CRC cells were assessed. Transcriptome analysis of HT-29 cells was performed using RNA sequencing. Metabolite profiling was conducted, and changes in biochemical parameters were evaluated using flow cytometry and biochemical analyses. RESULTS The combination index showed that HT-29 cells were the most responsive to the combined treatment, even with PIK3CA, B-RAF (V600E), and TP53 mutations. Moreover, ferroptosis was synergistically activated in the combined group of HT-29 in comparison to control. Furthermore, we observed an increase in OXPHOS metabolites, along with elevated intracellular and mitochondrial ROS, disruption of mitochondrial membrane potential, and greater levels of malondialdehyde (MDA) in the HT-29 combined group, which are the features of ferroptosis. Furthermore, ferroptosis induction was coupled with necroptosis, as indicated by RNA-sequencing data. Combination therapy inhibited cell migration and enhanced the immune response of HT-29 cells. CONCLUSIONS These findings suggest that PRO is promising as a potential adjuvant therapy in combination with CAP for the treatment of CRC. Only HT-29 cells with the B-RAF (V600E) mutation showed promising findings in this study.
Collapse
Affiliation(s)
- Shiekhah Mohammad Alzahrani
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
| | | | - Hind Ali Alkhatabi
- Department of Biological Science, College of Science, University of Jeddah, Jeddah P.O. Box 21589, Saudi Arabia
| | - Mohammad Abdullah Basabrain
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
| | - Peter Natesan Pushparaj
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
| |
Collapse
|
8
|
Perry NJS, Jhanji S, Poulogiannis G. Cancer Biology and the Perioperative Period: Opportunities for Disease Evolution and Challenges for Perioperative Care. Anesth Analg 2025; 140:846-859. [PMID: 39689009 DOI: 10.1213/ane.0000000000007328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Efforts to deconvolve the complex interactions of cancer cells with other components of the tumor micro- and macro-environment have exposed a common tendency for cancers to subvert systems physiology and exploit endogenous programs involved in homeostatic control of metabolism, immunity, regeneration, and repair. Many such programs are engaged in the healing response to surgery which, together with other abrupt biochemical changes in the perioperative period, provide an opportunity for the macroevolution of residual disease. This review relates contemporary perspectives of cancer as a systemic disease with the overlapping biology of host responses to surgery and events within the perioperative period. With a particular focus on examples of cancer cell plasticity and changes within the host, we explore how perioperative inflammation and acute metabolic, neuroendocrine, and immune dyshomeostasis might contribute to cancer evolution within this contextually short, yet crucially influential timeframe, and highlight potential therapeutic opportunities within to further optimize surgical cancer care and its long-term oncological outcomes.
Collapse
Affiliation(s)
- Nicholas J S Perry
- From the Signalling & Cancer Metabolism Team, Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Shaman Jhanji
- Department of Anaesthesia, Perioperative Medicine and Critical Care, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- Perioperative and Critical Care Outcomes Group, Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - George Poulogiannis
- From the Signalling & Cancer Metabolism Team, Division of Cancer Biology, The Institute of Cancer Research, London, UK
- Division of Computational and Systems Medicine, Department of Surgery & Cancer, Imperial College London, London, UK
| |
Collapse
|
9
|
Huang Z, Huang L, Zhang C, Chen G, Mai H. Blocking β2-AR and Inhibiting COX-2: A Promising Approach to Suppress OSCC Development. Int Dent J 2025; 75:807-816. [PMID: 39043526 PMCID: PMC11976482 DOI: 10.1016/j.identj.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
OBJECTIVES β2-adrenergic receptor (β2-AR) and cyclooxygenase-2 (COX-2) are overexpressed in various malignant tumours including oral squamous cell carcinoma (OSCC), suggesting that they may contribute to the development of OSCC. This study aims to investigate the potential synergistic effect of β2-AR blockade and COX-2 inhibition on suppressing the development of OSCC. METHODS Effects of blocking β2-AR and inhibiting COX-2 on migration and invasion of OSCC cells were detected by wound-healing assay and transwell invasion assay. Western blot and enzyme-linked immunosorbent assay (ELISA) were used to detect the expression of genes related to the progression of OSCC. In vivo, OSCC xenograft models were established to evaluate the effect of combined treatment on survival time, tumour size, and submandibular lymph node metastasis. Immunohistochemistry, Western blot, and ELISA were used to detect the expression of invasion and metastasis relative genes. RESULTS In vitro, blocking β2-AR or inhibiting COX-2 alone could suppress invasion and metastasis of OSCC cells, and suppression with combined treatment was more significant. Expression of genes related to invasion and metastasis, including EGFR, TGF-β1, IL-1β, MMP2, and VEGFA, were downregulated significantly, especially in the combined treatment group. In vivo, the combined treatment could significantly prolong survival time in tumour-bearing mice and inhibit the growth of tumours. Furthermore, submandibular lymph node metastasis was less in the combined treatment group, and expression of the abovementioned genes was also downregulated. CONCLUSIONS The combination of β2-AR blockade and COX-2 inhibition can significantly suppress the development of OSCC via downregulating EGFR, TGF-β1, IL-1β, MMP2, and VEGFA. Findings suggest that the combined use of a β2-AR blocker and a COX-2 inhibitor could be a promising adjuvant therapy in OSCC. Both drugs are commonly prescribed, and their safety and efficacy are well established. Their use in adjuvants in OSCC should therefore be promoted in clinical practice.
Collapse
Affiliation(s)
- Zeliu Huang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, China; Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, China
| | - Laifeng Huang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, China; Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, China
| | - Chong Zhang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, China; Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, China
| | - Guosheng Chen
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, China; Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, China
| | - Huaming Mai
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, China; Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, China.
| |
Collapse
|
10
|
Hadrevi J, Lu SSM, Järvholm LS, Palmqvist R, Olsson T, Harlid S, Van Guelpen B. Sick Leave due to Stress and Subsequent Cancer Risk, a Swedish National Registry Study of 516,678 Cancer Cases. Cancer Med 2025; 14:e70888. [PMID: 40247782 PMCID: PMC12006754 DOI: 10.1002/cam4.70888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/18/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND This study examined whether sick leave due to severe stress (stress leave) and duration of leave are associated with future cancer risk. METHODS We conducted a matched case-control study using complete-population data from Swedish national registers (2005 to 2018), including 516,678 primary cancer cases and 2,357,433 matched controls. Odds ratios (OR) were calculated by conditional logistic regression and adjusted for pre-specified confounders. RESULTS Stress leave of any duration, reported to the Swedish Social Insurance Register, was associated with a slightly increased cancer risk, with the highest risk estimate for 1-30 versus 0 days (adjusted OR 1.05, 95% CI 1.02-1.09). In men, a clear exposure-response trend was present. We observed increased risks of prostate cancer (adjusted OR for > 90 days: 1.10, 95% CI 1.01-1.20) and cervical cancer (adjusted OR for > 90 days: 1.11, 95% CI 1.05-1.17, including cancer in situ). In etiology-based analyses, a positive association was found for smoking-related cancers, and the risk relationship for non-cervical HPV-related cancers was similar to that for cervical cancer. Risk estimates were above one for several types of stress in relation to overall cancer risk, including an exposure-response trend for acute stress reactions (p-trend 4.0 × 10-4) but a null association for post-traumatic stress disorder. CONCLUSIONS Stress leave was associated with a modestly higher risk of cancer overall and prostate and cervical cancers specifically. Regardless of whether the link is biological or reflective of lifestyle mediators or for cervical cancer, lower participation in screening, these findings suggest a potential relevance of severe stress for cancer prevention.
Collapse
Affiliation(s)
- Jenny Hadrevi
- Section of Sustainable Health, Department of Global Health and EpidemiologyUmeå UniversityUmeåSweden
| | - Sai San Moon Lu
- Section of Sustainable Health, Department of Global Health and EpidemiologyUmeå UniversityUmeåSweden
- Section of Oncology, Department of Diagnostics and InterventionUmeå UniversityUmeåSweden
| | - Lisbeth Slunga Järvholm
- Section of Sustainable Health, Department of Global Health and EpidemiologyUmeå UniversityUmeåSweden
| | - Richard Palmqvist
- Section of Pathology, Department of Medical BiosciencesUmeå UniversityUmeåSweden
| | - Tommy Olsson
- Section of Medicine, Department of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Sophia Harlid
- Section of Oncology, Department of Diagnostics and InterventionUmeå UniversityUmeåSweden
| | - Bethany Van Guelpen
- Section of Oncology, Department of Diagnostics and InterventionUmeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicineUmeå UniversityUmeåSweden
| |
Collapse
|
11
|
Shen A, Liu F, Chen S, Huang K, Cao J, Shen C, Liu X, Yu L, Gu S, Zhao L, Li Y, Hu D, Huang J, Lu X, Gu D, Li J. Impact of resting heart rate and predicted cardiovascular risk on mortality in nearly 110,000 Chinese adults. Heart Rhythm 2025:S1547-5271(25)02252-0. [PMID: 40174737 DOI: 10.1016/j.hrthm.2025.03.1990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The association of mortality with resting heart rate (RHR) and predicted risk of incident cardiovascular disease (CVD) has not been comprehensively elucidated. OBJECTIVE The purpose of this study was to evaluate the individual and combined impacts of RHR and predicted CVD risk on mortality among Chinese adults. METHODS A total of 108,257 Chinese adults underwent follow-up during 1992 to 2015. The Cox proportional hazards model was used to assess the hazard ratio (HR) and 95% confidence interval (CI) of mortality associated with RHR and the predicted risk of incident CVD. RESULTS During an average of 8.53 years of follow-up, we identified 6267 nonaccidental deaths, including 2324 CVD deaths. High-RHR participants had higher HRs of nonaccidental (HR 1.58; 95% CI 1.47-1.70), CVD (HR 1.71; 95% CI 1.53-1.92), and non-CVD mortality (HR 1.50; 95% CI 1.37-1.64) than low-RHR participants. Compared to individuals with low CVD risk, those with high CVD risk demonstrated HRs of 1.67 (95% CI 1.51-1.83) and 3.90 (95% CI 3.29-4.61) for nonaccidental and CVD mortality, respectively. Mortality risk gradually increased with elevation of CVD risk and RHR, showing the greatest HRs of nonaccidental (HR 2.57; 95% CI 2.24-2.94) and CVD mortality (HR 7.16; 95% CI 5.52-9.29) among participants with both high CVD risk and high RHR. CONCLUSION Our findings demonstrated that RHR and CVD risk are independently associated with mortality, and integrating them potentially could refine risk stratification of mortality. The study highlighted their important role in personalized strategies for primordial and primary prevention.
Collapse
Affiliation(s)
- Anna Shen
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Fangchao Liu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufeng Chen
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Keyong Huang
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Cao
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chong Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoqing Liu
- Division of Epidemiology, Guangdong Provincial People's Hospital and Cardiovascular Institute, Guangzhou, China
| | - Ling Yu
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou, China
| | - Shujun Gu
- Department of Chronic Disease Control and Prevention, Changshu Center for Disease Control and Prevention, Changshu, China
| | - Liancheng Zhao
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Li
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongsheng Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, China; Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jianfeng Huang
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangfeng Lu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Dongfeng Gu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China; School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jianxin Li
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
12
|
Liang X, Li P, Qin Y, Mo Y, Chen D. Beta-adrenergic receptor blockers improve survival in patients with advanced non-small cell lung cancer combined with hypertension undergoing radiotherapy. Sci Rep 2025; 15:10702. [PMID: 40155651 PMCID: PMC11953261 DOI: 10.1038/s41598-025-93205-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
Hypertension (HTN) is prevalent in non-small cell lung cancer (NSCLC) patients, yet the cardioprotective and survival benefits of β-adrenergic blockers during radiotherapy (RT) remain underexplored. We analyzed data from a Chinese clinical cohort of 750 patients with stage IIIA to IIIB NSCLC and HTN receiving RT between 2014 and 2018. The findings were further validated using data from the NHANES database. In Chinese clinical cohort, β-adrenergic blockers were associated with improved OS (β-adrenergic blockers: median overall survival (mOS) 17.64 months, 95% CI, 15.95-19.33; no β-adrenergic blockers: mOS 13.16 months, 95% CI, 12.62-13.70; p < 0.0001) and PFS (β-adrenergic blockers: median progression-free survival (mPFS) 7.50 months, 95% CI, 6.50-8.50; without β-adrenergic blockers: mPFS 4.91 months, 95% CI, 4.53-5.31; p < 0.0001). Simultaneously, in the NHANES database, the utilization of β-adrenergic blockers exhibited no discernible impact on OS within the entire tumor population, as evidenced by the Kaplan-Meier curve, which revealed no statistically significant difference between the two groups (p = 0.254). β-adrenergic blockers may improve OS and PFS in patients with HTN and NSCLC undergoing RT. β-adrenergic blockers show potential and warrant further investigation in the context of RT.
Collapse
Affiliation(s)
- Xinyi Liang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261000, Shandong, People's Republic of China
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan, 250000, Shandong, People's Republic of China
| | - Pengwei Li
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan, 250000, Shandong, People's Republic of China
| | - Yiwei Qin
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan, 250000, Shandong, People's Republic of China
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - You Mo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, 515000, Guangdong, People's Republic of China.
| | - Dawei Chen
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan, 250000, Shandong, People's Republic of China.
| |
Collapse
|
13
|
MAFFEZZOLI MICHELE, GIUDICE GIULIACLAIRE, IOVANE GIACOMO, MANINI MARTINA, RAPACCHI ELENA, CARUSO GIUSEPPE, SIMONI NICOLA, FERRETTI STEFANIA, PULIATTI STEFANO, CAMPOBASSO DAVIDE, BUTI SEBASTIANO. The effect of concomitant drugs on oncological outcomes in patients treated with immunotherapy for metastatic urothelial carcinoma: a narrative review. Oncol Res 2025; 33:741-757. [PMID: 40191722 PMCID: PMC11964881 DOI: 10.32604/or.2024.057278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/04/2024] [Indexed: 04/09/2025] Open
Abstract
Background immune checkpoint inhibitors (ICIs) have revolutionized the treatment of metastatic urothelial carcinoma (mUC), significantly improving survival outcomes. However, a subset of patients do not respond to ICIs, prompting research into potential predictive factors. Commonly prescribed medications such as corticosteroids, proton-pump inhibitors (PPIs), antibiotics (Abs), antihypertensives, and analgesics may influence ICI effectiveness. Methods we conducted a literature search on PubMed to investigate the impact of concomitant medications on the outcomes of patients with mUC, treated with ICIs. We selected the most relevant studies and performed a narrative review. Results corticosteroids, PPIs and Abs have been associated with reduced survival in ICI-treated patients, including those with mUC. In contrast, antihypertensive agents like renin-angiotensin system inhibitors and beta-blockers may enhance ICI efficacy, though evidence remains inconclusive. The impact of other medications, such as statins, metformin, and analgesics, on ICI outcomes is less clear, with some data suggesting a detrimental impact on immune response. Conclusions this narrative review synthesizes current evidence on how concomitant medications affect outcomes in mUC patients treated with ICIs.
Collapse
Affiliation(s)
- MICHELE MAFFEZZOLI
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| | - GIULIA CLAIRE GIUDICE
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| | - GIACOMO IOVANE
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| | - MARTINA MANINI
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| | - ELENA RAPACCHI
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
| | - GIUSEPPE CARUSO
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
| | - NICOLA SIMONI
- Radiotherapy Unit, University Hospital of Parma, Parma, 43126, Italy
| | - STEFANIA FERRETTI
- Department of Urology, University of Modena and Reggio Emilia, Modena, 41124, Italy
| | - STEFANO PULIATTI
- Department of Urology, University of Modena and Reggio Emilia, Modena, 41124, Italy
| | | | - SEBASTIANO BUTI
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| |
Collapse
|
14
|
Jiauddin M, Reddy K, Ravi HP, Ramachandran B. Druggable upregulated proteins in EWS-FLI-driven Ewing sarcoma as emerging new therapeutic targets. Am J Transl Res 2025; 17:1580-1603. [PMID: 40225989 PMCID: PMC11982847 DOI: 10.62347/ymeu1808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
Ewing sarcoma (ES) is a highly aggressive soft tissue tumor that primarily affects the long bones of children and young adults. It is distinguished by a characteristic chromosomal translocation between the Ewing sarcoma breakpoint region 1 (EWS) gene and the erythroblast transformation-specific (ETS) family of genes, most commonly resulting in the EWS-friend leukemia integration 1 (EWS-FLI1) fusion gene. This translocation is observed in approximately 80%-85% of ES cases. This fusion gene encodes a non-physiological chimeric fusion protein that plays a central role in tumorigenesis by interacting with numerous partner proteins. Several studies have demonstrated the tumorigenic potential of the EWS-FLI1 protein when transfected into non-cancer cell lines. However, targeting EWS-FLI1 directly remains a significant challenge, as no drug to date has been reported to bind to and inhibit its activity effectively. An alternative therapeutic strategy involves targeting key overexpressed protein complexes implicated in ES tumorigenesis, many of which may be downstream interacting partners of EWS-FLI1. This review explores emerging protein targets as potential therapeutic avenues in ES treatment.
Collapse
Affiliation(s)
- Moinuddin Jiauddin
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| | - Kirtana Reddy
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| | - Hashiya Preeya Ravi
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| | - Balaji Ramachandran
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| |
Collapse
|
15
|
Wang Y, Anesi JC, Panicker IS, Cook D, Bista P, Fang Y, Oqueli E. Neuroimmune Interactions and Their Role in Immune Cell Trafficking in Cardiovascular Diseases and Cancer. Int J Mol Sci 2025; 26:2553. [PMID: 40141195 PMCID: PMC11941982 DOI: 10.3390/ijms26062553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Sympathetic nerves innervate bone marrow and various immune organs, where norepinephrine-the primary sympathetic neurotransmitter-directly interacts with immune cells that express adrenergic receptors. This article reviewed the key molecular pathways triggered by sympathetic activation and explored how sympathetic activity influences immune cell migration. Norepinephrine serves as a chemoattractant for monocytes, macrophages, and stem cells, promoting the migration of myeloid cells while inhibiting the migration of lymphocytes at physiological concentrations. We also examined the role of immune cell infiltration in cardiovascular diseases and cancer. Evidence suggests that sympathetic activation increases myeloid cell infiltration into target tissues across various cardiovascular diseases, including atherosclerosis, hypertension, cardiac fibrosis, cardiac hypertrophy, arrhythmia, myocardial infarction, heart failure, and stroke. Conversely, inhibiting sympathetic activity may serve as a potential therapeutic strategy to treat these conditions by reducing macrophage infiltration. Furthermore, sympathetic activation promotes macrophage accumulation in cancer tissues, mirroring its effects in cardiovascular diseases, while suppressing T lymphocyte infiltration into cancerous sites. These changes contribute to increased cancer growth and metastasis. Thus, inhibiting sympathetic activation could help to protect against cancer by enhancing T cell infiltration and reducing macrophage presence in tumors.
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Jack C. Anesi
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Indu S. Panicker
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Darcy Cook
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Prapti Bista
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Yan Fang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Ernesto Oqueli
- Cardiology Department, Grampians Health Ballarat, Ballarat, VIC 3353, Australia
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC 3217, Australia
| |
Collapse
|
16
|
Komine C, Sohda M, Yokobori T, Shioi I, Ozawa N, Shibasaki Y, Nakazawa N, Osone K, Shiraishi T, Okada T, Sano A, Sakai M, Ogawa H, Kaira K, Shirabe K, Saeki H. Impact of Tumoral β2-Adrenergic Receptor Expression on Chemotherapeutic Response and Prognosis in Patients with Advanced Colorectal Cancer. Ann Surg Oncol 2025; 32:1913-1924. [PMID: 39341920 DOI: 10.1245/s10434-024-16195-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND The β2-adrenergic receptor (β2-AR) is a therapeutic target for circulatory agonists and exhibits oncogenic activity in several cancers. However, its role in advanced colorectal cancer (CRC) treated using chemotherapy remains unclear. We investigated the potential of β2-AR as a novel chemosensitivity marker and therapeutic target in inoperable CRC. METHODS β2-AR expression was evaluated immunohistochemically in 80 advanced or recurrent CRC cases for which untreated resected specimens were available before systemic chemotherapy implementation. We assessed the relationship among β2-AR protein expression, clinicopathological factors, therapeutic response, and prognosis. Furthermore, we evaluated the significance of β2-AR as an in vitro and in vivo therapeutic target using CRC cell lines and a CRC xenograft model treated with the β-blocker, propranolol, and other anticancer agents. RESULTS High tumoral β2-AR expression was associated with shorter progression-free survival and chemotherapeutic resistance in patients treated with oxaliplatin-based regimens and bevacizumab-based regimens. We found no synergistic effect between propranolol and oxaliplatin. However, combined administration of propranolol and bevacizumab induced significant tumor shrinkage in the CRC xenograft model. CONCLUSIONS β2-AR is a possible biomarker for chemosensitivity and prognosis in advanced CRC. Repositioning existing β-blockers could be beneficial for treating CRC resistant to existing treatment regimens.
Collapse
MESH Headings
- Humans
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Receptors, Adrenergic, beta-2/metabolism
- Animals
- Female
- Prognosis
- Male
- Mice
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/metabolism
- Propranolol/administration & dosage
- Propranolol/pharmacology
- Oxaliplatin
- Survival Rate
- Middle Aged
- Xenograft Model Antitumor Assays
- Drug Resistance, Neoplasm
- Aged
- Bevacizumab/administration & dosage
- Tumor Cells, Cultured
- Follow-Up Studies
- Mice, Nude
- Mice, Inbred BALB C
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Organoplatinum Compounds/administration & dosage
- Cell Proliferation/drug effects
- Adult
Collapse
Affiliation(s)
- Chika Komine
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Makoto Sohda
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan.
| | - Takehiko Yokobori
- Research Program for Omics-Based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan.
| | - Ikuma Shioi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Naoya Ozawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Yuta Shibasaki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Nobuhiro Nakazawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Katsuya Osone
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Takuya Shiraishi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Takuhisa Okada
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Akihiko Sano
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Makoto Sakai
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Hiroomi Ogawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Kyoichi Kaira
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama University Hospital, Hidaka, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Hiroshi Saeki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| |
Collapse
|
17
|
Chang Y, Zhou H, Ren Y, Zhang J, Sun L, Du M, Zhao J, Chu H, Zhao Y. Identifying multi-target drugs for prostate cancer using machine learning-assisted transcriptomic analysis. J Biomol Struct Dyn 2025; 43:2109-2119. [PMID: 38102880 DOI: 10.1080/07391102.2023.2294168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/30/2023] [Indexed: 12/17/2023]
Abstract
Prostate cancer is a leading cause of cancer death in men, and the development of effective treatments is of great importance. This study explored to identify the candidate drugs for prostate cancer by transcriptomic data and CMap database analysis. After integrating the results of omics analysis, bisoprolol is confirmed as a promising drug. Moreover, cell experiment reveals its potential inhibitory effect on the proliferation of prostate cancer cells. Importantly, machine learning methods are employed to predict the targets of bisoprolol, and the dual-target ADRB3 and hERG are explored by dynamic simulation. The findings of this study demonstrate the potential of bisoprolol as a multi-target drug for prostate cancer treatment and the feasibility of using beta-adrenergic receptor inhibitors in prostate cancer treatment. In addition, the proposed research approach is promising for discovering potential drugs for cancer treatment by leveraging the concept of drug side effects leading to anticancer effects. Further research is necessary to investigate the pharmacological action, potential toxicity, and underlying mechanisms of bisoprolol in treating prostate cancer with ADRB3.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yibin Chang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Hongmei Zhou
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuxiang Ren
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Jiaqi Zhang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Lei Sun
- College of Medical Devices, Shenyang Pharmaceutical University, Shenyang, China
| | - Minghui Du
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Jian Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Huiying Chu
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yongshan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
18
|
Earnhardt-San AL, Baker EC, Cilkiz KZ, Cardoso RC, Ghaffari N, Long CR, Riggs PK, Randel RD, Riley DG, Welsh TH. Evaluation of Prenatal Transportation Stress on DNA Methylation (DNAm) and Gene Expression in the Hypothalamic-Pituitary-Adrenal (HPA) Axis Tissues of Mature Brahman Cows. Genes (Basel) 2025; 16:191. [PMID: 40004522 PMCID: PMC11855312 DOI: 10.3390/genes16020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The experience of prenatal stress results in various physiological disorders due to an alteration of an offspring's methylome and transcriptome. The objective of this study was to determine whether PNS affects DNA methylation (DNAm) and gene expression in the stress axis tissues of mature Brahman cows. Methods: Samples were collected from the paraventricular nucleus (PVN), anterior pituitary (PIT), and adrenal cortex (AC) of 5-year-old Brahman cows that were prenatally exposed to either transportation stress (PNS, n = 6) or were not transported (Control, n = 8). The isolated DNA and RNA samples were, respectively, used for methylation and RNA-Seq analyses. A gene ontology and KEGG pathway enrichment analysis of each data set within each sample tissue was conducted with the DAVID Functional Annotation Tool. Results: The DNAm analysis revealed 3, 64, and 99 hypomethylated and 2, 93, and 90 hypermethylated CpG sites (FDR < 0.15) within the PVN, PIT, and AC, respectively. The RNA-Seq analysis revealed 6, 25, and 5 differentially expressed genes (FDR < 0.15) in the PVN, PIT, and AC, respectively, that were up-regulated in the PNS group relative to the Control group, as well as 24 genes in the PIT that were down-regulated. Based on the enrichment analysis, several developmental and cellular processes, such as maintenance of the actin cytoskeleton, cell motility, signal transduction, neurodevelopment, and synaptic function, were potentially modulated. Conclusions: The methylome and transcriptome were altered in the stress axis tissues of mature cows that had been exposed to prenatal transportation stress. These findings are relevant to understanding how prenatal experiences may affect postnatal neurological functions.
Collapse
Affiliation(s)
- Audrey L. Earnhardt-San
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA
| | - Emilie C. Baker
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Kubra Z. Cilkiz
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Rodolfo C. Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Noushin Ghaffari
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Charles R. Long
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Penny K. Riggs
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Ronald D. Randel
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| |
Collapse
|
19
|
Harris AR, Pichardo CM, Franklin J, Liu H, Wooten W, Panigrahi G, Lawrence WR, Pichardo MS, Jenkins BD, Dorsey TH, Ioffe OB, Yfantis HG, Agurs-Collins T, Ambs S. Multilevel Stressors and Systemic and Tumor Immunity in Black and White Women With Breast Cancer. JAMA Netw Open 2025; 8:e2459754. [PMID: 39951265 PMCID: PMC11829235 DOI: 10.1001/jamanetworkopen.2024.59754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/05/2024] [Indexed: 02/17/2025] Open
Abstract
Importance The mechanisms through which chronic stressors may be associated with tumor biologic characteristics, immune response, and health disparities remain insufficiently understood. Objective To investigate the proteomic, transcriptomic, and genomic effects associated with multilevel chronic stressors (perceived stress, perceived inadequate social support, perceived racial and ethnic discrimination, and neighborhood deprivation) in Black and White women with breast cancer. Design, Setting, and Participants This cross-sectional study was conducted from February 28, 2012, to September 5, 2023, in which blood samples, breast tumors, and adjacent noncancerous tissues were collected from women with breast cancer. Participants, recruited at 2 Baltimore, Maryland, hospitals, completed demographic and psychosocial questionnaires. Data analysis was conducted from September 2023 to April 2024. Exposures Perceived stress, perceived social support, perceived racial and ethnic discrimination, and the 2010 census tract-level neighborhood deprivation index, in which scores range from -2.51 to 6.77, with higher scores indicating greater deprivation. Main Outcomes and Measures The main outcomes included levels of 92 circulating immune-oncologic markers and associated biologic pathways, tumor immune cell profiles, breast tissue gene expression, and tumor mutational burden. Data were analyzed using covariate-adjusted linear regression modeling for continuous outcomes with effect estimates presented as β values with 95% CIs. Results The study included 121 women with breast cancer (mean [SD] age, 56.27 [12.62] years), of whom 56 (46.3%) were Black, and 65 (53.7%) were White. The analytic subsample sizes included 117 blood samples, 48 breast tumors, and 41 adjacent noncancerous tissues. Levels of perceived stress and social support were comparable by race, while Black women resided in more socioeconomically deprived neighborhoods (mean [SD] neighborhood deprivation index, 2.28 [2.30] for Black women compared with -0.22 [2.01] for White women). Greater perceived social support was associated with more favorable immune-stimulatory changes (eg, increased serum IL-5 [β, 0.06 (95% CI, 0.02-0.10); P = .003] and activated natural killer cells in noncancerous breast tissue of Black women [β, 0.11 (95% CI, 0.04-0.17); P = .002). Higher levels of perceived stress, exposure to discrimination, and neighborhood deprivation were associated with systemic inflammation (eg, serum IL-6 with both perceived stress [β, 0.04 (95% CI, 0.01-0.07); P = .006] and discrimination [β, 0.69 (95% CI, 0.15-1.23); P = .01]); deleterious immune cell profiles (eg, tumor-associated M2 macrophages with discrimination [β, 0.82 (95% CI, 0.14-1.51); P = .02]); and aggressive tumor biologic characteristics. Race-stratified analyses uncovered distinct immunologic features in Black women associated with stressors, including chemotaxis with stress (β, 0.28 [95% CI, 0.001-0.56]; P = .049) and immune suppression with stress (β, 0.37 [95% CI, -0.002 to 0.75]; P = .05) at the systemic level and increased tumor-associated myeloid cells (monocytes and M1 and M2 macrophages) at the tissue level. Perceived stress was associated with elevated tumor mutational burden (β, 0.02 [95% CI, 0.01-0.04]; P = .04). Conclusions and Relevance The findings of this cross-sectional study of Black and White women with breast cancer suggest that perceived stress, perceived inadequate social support, perceived racial and ethnic discrimination, and neighborhood deprivation were associated with deleterious alterations to the systemic and tumor immune environment, particularly for Black women. Understanding biology as a possible mediator of cancer health disparities may inform prevention and public health interventions.
Collapse
Affiliation(s)
- Alexandra R. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Catherine M. Pichardo
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Jamirra Franklin
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - William Wooten
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center Biostatistics Shared Service, Baltimore
| | - Gatikrushna Panigrahi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wayne R. Lawrence
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | | | - Brittany D. Jenkins
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Tiffany H. Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Olga B. Ioffe
- Department of Pathology, University of Maryland School of Medicine, Baltimore
| | - Harry G. Yfantis
- Pathology and Laboratory Medicine, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
20
|
Taylor MR, Bradford MC, Zhou C, Fladeboe KM, Wittig JF, Baker KS, Yi‐Frazier JP, Rosenberg AR. Heart Rate Variability as a Digital Biomarker in Adolescents and Young Adults Receiving Hematopoietic Cell Transplantation. Cancer Med 2025; 14:e70609. [PMID: 39981705 PMCID: PMC11843223 DOI: 10.1002/cam4.70609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 02/22/2025] Open
Abstract
INTRODUCTION Adolescents and young adults (AYAs) receiving hematopoietic cell transplantation (HCT) are at high risk for poor psychosocial outcomes. Heart rate variability (HRV), a surrogate for autonomic nervous system activity, is a promising digital biomarker that has been linked to important outcomes. The objectives of this study were to prospectively describe the trajectory of HRV among AYAs receiving HCT and explore the association between HRV and patient-reported outcomes (PROs). METHODS This was a multi-site study embedded in a randomized trial among AYAs receiving HCT (NCT03640325). We collected sequential 24-h HRV metrics, including the standard deviation of normal-to-normal beats (SDNN), root-mean-square of successive differences (RMSSD), as well as frequency domain measures. PRO surveys queried anxiety, depression, quality of life, hope, and resilience at baseline and 3 months. We summarized outcomes using descriptive statistics, and Pearson correlation coefficients were used to examine the relationship between HRV and PROs. RESULTS Thirty-nine HRV recordings were collected from n = 16 participants aged 12-21 years. There was a moderately strong correlation between inferior baseline HRV and higher anxiety and depression (anxiety: r = -0.35 (p = 0.18) for SDNN, r = -0.47 (p = 0.07) for RMSSD; depression: r = -0.26 (p = 0.34) for SDNN, r = -0.39 (p = 0.14) for RMSSD). Among participants with elevated baseline anxiety, higher HRV suggested greater improvement in anxiety over time (r = -0.66 (p = 0.08) for SDNN, r = -0.31 (p = 0.45) for RMSSD). CONCLUSIONS There was a correlation between HRV and PROs in this study, and among those with elevated anxiety, HRV predicted improvement over time. Digital biomarkers may augment behavioral intervention design and implementation.
Collapse
Affiliation(s)
- Mallory R. Taylor
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashingtonUSA
- Ben Towne Center for Childhood Cancer and Blood Disorders ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Miranda C. Bradford
- Biostatistics Epidemiology and Analytics in Research CoreSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Chuan Zhou
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashingtonUSA
- Center for Child Health, Behavior, and DevelopmentSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Kaitlyn M. Fladeboe
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashingtonUSA
- Ben Towne Center for Childhood Cancer and Blood Disorders ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Jorie F. Wittig
- University of Washington School of MedicineSeattleWashingtonUSA
| | - K. Scott Baker
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashingtonUSA
- Clinical Research DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Joyce P. Yi‐Frazier
- Department of Psychosocial Oncology and Palliative CareDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Abby R. Rosenberg
- Department of Psychosocial Oncology and Palliative CareDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of PediatricsBoston Children's HospitalBostonMassachusettsUSA
- Department of PediatricsHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
21
|
Elebyary TT, Sultan AA, Abu-Risha SE, El Maghraby GM, Amin M. Bilosomal Co-Encapsulated Tamoxifen and Propranolol for Potentiated Anti-Breast Cancer Efficacy: In Vitro and In Vivo Investigation. Pharmaceutics 2025; 17:123. [PMID: 39861770 PMCID: PMC11768151 DOI: 10.3390/pharmaceutics17010123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Tamoxifen (TAM) is an anti-breast cancer drug suffering from acquired resistance development, prompting cancer relapse. Propranolol (PRO)'s repurposing for cancer therapy has gained interest. This work aimed to investigate combined TAM/PRO therapy for potentiating the anti-breast cancer activity of TAM. The work probed bilosomes versus standard noisome for simultaneous oral and intratumor delivery of TAM and PRO. Methods: Bilosomes comprising Span60, cholesterol, and increasing concentrations of bile salts were prepared together with bile salts containing free standard niosomes. The vesicular size and morphology were characterized. The entrapment and release efficiencies of TAM and PRO from the tailored vesicles were determined. The in vivo investigations of anti-tumor activity of TAM with or without PRO employed the solid Ehrlich carcinoma model. Results: The vesicles of all fabricated dispersions were spherical and negatively charged, with a size ranging from 104 to 182 nm. The entrapment efficiency depended on the nature of the drug, recording values ranging from 87.5% to 97.8% for TAM and from 31.0% to 46.8% for PRO. Incorporation of bile salts into vesicles increased TAM and PRO release compared to standard niosomes. Oral administration of combined TAM/PRO bilosomes showed a significant reduction in tumor growth volume compared to that recorded following naked drug administration. Histopathological investigations reflected a significant decline in tumor giant cells and mitotic figures, implying the in vivo capability of the TAM/PRO combination to interfere with cancer cell proliferation and persistence. Conclusions: The overall results demonstrated the impact of repurposed PRO to enhance the anti-breast cancer activity of TAM when both were co-encapsulated into bilosomes.
Collapse
Affiliation(s)
- Toka T. Elebyary
- Department of Pharmaceutical Technology, Faculty of pharmacy, Tanta University, Tanta 31527, Egypt; (T.T.E.); (G.M.E.M.)
- Educational Hospital, Tanta University, Tanta 31527, Egypt
| | - Amal A. Sultan
- Department of Pharmaceutical Technology, Faculty of pharmacy, Tanta University, Tanta 31527, Egypt; (T.T.E.); (G.M.E.M.)
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 39911, Saudi Arabia
| | - Sally E. Abu-Risha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt;
| | - Gamal M. El Maghraby
- Department of Pharmaceutical Technology, Faculty of pharmacy, Tanta University, Tanta 31527, Egypt; (T.T.E.); (G.M.E.M.)
- Faculty of Pharmacy, Alsalam University, Tanta 31527, Egypt
| | - Manna Amin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| |
Collapse
|
22
|
Park HJ, Lee SC, Park SH. Norepinephrine stimulates M2 macrophage polarization via β2-adrenergic receptor-mediated IL-6 production in breast cancer cells. Biochem Biophys Res Commun 2024; 741:151087. [PMID: 39616942 DOI: 10.1016/j.bbrc.2024.151087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/11/2024]
Abstract
Previous studies have demonstrated that norepinephrine (NE) released during chronic stress promotes breast cancer (BC) metastasis via adrenergic receptors (ARs). However, the effect of NE on tumor-associated macrophage polarization and the underlying mechanisms remain largely unknown. In this study, we aimed to investigate the influence of NE on M2 macrophage polarization, with a particular focus on the crosstalk between macrophages and BC cells. Our results demonstrated that, although NE alone did not directly induce the expression of M2 macrophage markers, conditioned medium from NE-treated MDA-MB-231 human BC cells (NE CM) significantly promoted M2 macrophage polarization in THP-1 macrophages. We found that NE stimulated IL-6 production in MDA-MB-231 cells via β2-AR/NF-κB pathway, which activated STAT3 in THP-1 cells to induce M2 macrophage polarization. NE failed to induce IL-6 production and NF-κB activation when ADRB2 was knocked down in MDA-MB-231 cells. Furthermore, ADRB2 knockdown in cancer cells suppressed NE CM-induced M2 macrophage polarization, as well as M2 macrophage-induced cancer cell migration. Taken together, our results suggest that NE stimulates M2 macrophage polarization by inducing IL-6 secretion from BC cells through a β2-AR-dependent mechanism, which subsequently promotes cancer cell migration. Targeting β2-AR may represent a promising strategy to prevent chronic stress-induced BC metastasis.
Collapse
Affiliation(s)
- Hyun-Ji Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan, 47227, Republic of Korea
| | - Su-Chan Lee
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Shin-Hyung Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan, 47227, Republic of Korea.
| |
Collapse
|
23
|
Molnár AÁ, Birgés K, Surman A, Merkely B. The Complex Connection Between Myocardial Dysfunction and Cancer Beyond Cardiotoxicity: Shared Risk Factors and Common Molecular Pathways. Int J Mol Sci 2024; 25:13185. [PMID: 39684895 DOI: 10.3390/ijms252313185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiovascular diseases and cancer represent the largest disease burden worldwide. Previously, these two conditions were considered independent, except in terms of cardiotoxicity, which links cancer treatment to subsequent cardiovascular issues. However, recent studies suggest that there are further connections between cancer and heart disease beyond cardiotoxicity. It has been revealed that myocardial dysfunction may promote carcinogenesis, indicating that additional common pathophysiological mechanisms might be involved in the relationship between cardiology and oncology, rather than simply a connection through cardiotoxic effects. These mechanisms may include shared risk factors and common molecular pathways, such as persistent inflammation and neurohormonal activation. This review explores the connection between myocardial dysfunction and cancer, emphasizing their shared risk factors, similar biological mechanisms, and causative factors like cardiotoxicity, along with their clinical implications.
Collapse
Affiliation(s)
| | - Kristóf Birgés
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Adrienn Surman
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| |
Collapse
|
24
|
Dong ZK, Wang YF, Li WP, Jin WL. Neurobiology of cancer: Adrenergic signaling and drug repurposing. Pharmacol Ther 2024; 264:108750. [PMID: 39527999 DOI: 10.1016/j.pharmthera.2024.108750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Cancer neuroscience, as an emerging converging discipline, provides us with new perspectives on the interactions between the nervous system and cancer progression. As the sympathetic nervous system, in particular adrenergic signaling, plays an important role in the regulation of tumor activity at every hierarchical level of life, from the tumor cell to the tumor microenvironment, and to the tumor macroenvironment, it is highly desirable to dissect its effects. Considering the far-reaching implications of drug repurposing for antitumor drug development, such a large number of adrenergic receptor antagonists on the market has great potential as one of the means of antitumor therapy, either as primary or adjuvant therapy. Therefore, this review aims to summarize the impact of adrenergic signaling on cancer development and to assess the status and prospects of intervening in adrenergic signaling as a therapeutic tool against tumors.
Collapse
Affiliation(s)
- Zi-Kai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Yong-Fei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Wei-Ping Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Urology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Wei-Lin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
25
|
Taylor MR, Cole SW, Bradford MC, Zhou C, Fladeboe KM, Knight JM, Baker KS, Yi-Frazier JP, Rosenberg AR. Resilience Intervention Improves Stress-Related Gene Expression in Adolescent and Young Adult HCT Recipients. Transplant Cell Ther 2024; 30:1209.e1-1209.e7. [PMID: 39303988 DOI: 10.1016/j.jtct.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Overactivation of the stress response can influence cancer outcomes through immune-related pathways. Adolescents and young adults (AYAs) undergoing hematopoietic cell transplantation (HCT) are at risk for poor outcomes, yet there are limited behavioral interventions and no psychosocial biomarker data for this population. The Conserved Transcriptional Response to Adversity (CTRA) is an inflammation-related pattern observed in conditions of heightened stress and is associated with HCT outcomes. The objective of the current study was to explore the CTRA gene regulatory impact of Promoting Resilience in Stress Management (PRISM) intervention among AYAs receiving HCT. We hypothesized that patients who received the intervention would have favorable gene expression signatures compared to those in the control arm. This was an ancillary study within a randomized trial testing the PRISM intervention on psychosocial outcomes among AYAs aged 12 to 24 years receiving HCT (NCT03640325). CTRA was quantified through genome-wide transcriptional profiles obtained from whole blood collected at baseline, 1-, and 3-month post-HCT. Group differences in CTRA gene expression were estimated using mixed-effect linear models. There were no baseline group differences in CTRA expression, but PRISM participants showed a greater decline in CTRA at 1 month compared to controls (β -0.301 ± SE 0.114, P = .016), even when controlling for demographic (Group × Time interaction: F(2, 18) = 7.41, P = .004; β -0.386 ± 0.127, P = .007) and clinical covariates (Group × Time interaction: F(2, 20) = 7.03, P = .005; β -0.480 ± 0.144, P = .003). These differences were not detectable at 3 months (β -0.147 ± SE 0.120, P = .235). There was a change in stress-related gene expression among AYAs randomized to a psychosocial intervention. The stress-inflammation axis may be a targetable pathway in the AYA HCT population.
Collapse
Affiliation(s)
- Mallory R Taylor
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington; Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington.
| | - Steve W Cole
- Departments of Psychiatry & Biobehavioral Sciences and Medicine, UCLA School of Medicine, Los Angeles, California
| | - Miranda C Bradford
- Biostatistics Epidemiology and Analytics in Research Core, Seattle Children's Research Institute, Seattle, Washington
| | - Chuan Zhou
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington; Center for Child Health, Behavior, and Development, Seattle Children's Research Institute, Seattle, Washington
| | - Kaitlyn M Fladeboe
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington; Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington
| | - Jennifer M Knight
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - K Scott Baker
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington; Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington
| | - Joyce P Yi-Frazier
- Department of Psychosocial Oncology and Palliative Care, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Abby R Rosenberg
- Department of Psychosocial Oncology and Palliative Care, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
26
|
Dongdem JT, Etornam AE, Beletaa S, Alidu I, Kotey H, Wezena CA. The β 3-Adrenergic Receptor: Structure, Physiopathology of Disease, and Emerging Therapeutic Potential. Adv Pharmacol Pharm Sci 2024; 2024:2005589. [PMID: 39640497 PMCID: PMC11620816 DOI: 10.1155/2024/2005589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
The discovery and characterization of the signal cascades of the β-adrenergic receptors have made it possible to effectively target the receptors for drug development. β-Adrenergic receptors are a class A rhodopsin type of G protein-coupled receptors (GPCRs) that are stimulated mainly by catecholamines and therefore mediate diverse effects of the parasympathetic nervous system in eliciting "fight or flight" type responses. They are detectable in several human tissues where they control a plethora of physiological processes and therefore contribute to the pathogenesis of several disease conditions. Given the relevance of the β-adrenergic receptor as a molecular target for many pathological conditions, this comprehensive review aims at providing an in-depth exploration of the recent advancements in β3-adrenergic receptor research. More importantly, we delve into the prospects of the β3-adrenergic receptor as a therapeutic target across a variety of clinical domains.
Collapse
Affiliation(s)
- Julius T. Dongdem
- Department of Chemical Pathology, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Axandrah E. Etornam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Solomon Beletaa
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Issah Alidu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Hassan Kotey
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Cletus A. Wezena
- Department of Microbiology, Faculty of Biosciences, University for Development Studies, Tamale, Northern Region, Ghana
| |
Collapse
|
27
|
Mikolaskova I, Zvarik M, Szaboova K, Tibenska E, Durmanova V, Suchankova M, Kollarik B, Hesko P, Palacka P, Bucova M, Hunakova L. Association of Sympathovagal Imbalance with Increased Inflammation and Impaired Adaptive Immunity in Bladder Cancer Patients. Int J Mol Sci 2024; 25:12765. [PMID: 39684475 DOI: 10.3390/ijms252312765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Stress responses can impact bladder cancer (BC) outcomes via immune-inflammatory pathway modulation. This study explores heart rate variability (HRV) associations with serum immune-inflammatory biomarkers, blood count inflammatory markers, and psychosocial self-report measures in patients versus healthy controls. The TREM-1 and TREM-2 expressions on peripheral blood monocytes were analysed via flow cytometry; serum inflammatory biomarkers by ELISA; HRV (5-min ECG) pre-tumour resection; blood counts by haematology analyser; and psychosocial factors by validated questionnaires. Patients exhibited altered immune-inflammatory profiles with increased TREM-1/TREM-2, sTREM-1, sTREM-1/sTREM-2 ratio, BDNF, MCP-1, and NLR, and reduced IFN-γ, IL-10, LMR, and PMR. HRV analysis indicated sympathetic dominance (SNS, Stress indices, ACmod) and reduced parasympathetic modulation (PNS index, SDNN, RMSSD, 2UV%, DCmod, SD1). Sympathetic HRV indices correlated positively with sTREM-1, sTREM-1/sTREM-2 ratio, fractalkine, and inflammatory markers (SII, NLR, PLR) and negatively with parasympathetic HRV indices-correlations absent in controls. Only in patients, reduced physical function and social support, and higher anxiety, depression, and fatigue, associated positively with sympathetic HRV indices and inflammatory markers. This study links immune-inflammatory markers, HRV parameters, and psychosocial factors in BC, suggesting that immune and autonomic variations may relate to unfavourable outcomes. Incorporating these assessments could help tailor more personalised treatment strategies for BC patients.
Collapse
Affiliation(s)
- Iveta Mikolaskova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08 Bratislava, Slovakia
| | - Milan Zvarik
- Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Computer Science, Comenius University in Bratislava, Mlynska dolina F1, 842 48 Bratislava, Slovakia
| | - Kinga Szaboova
- Medirex, s.r.o., Galvaniho 17/C, 820 16 Bratislava, Slovakia
| | - Elena Tibenska
- Medirex, s.r.o., Galvaniho 17/C, 820 16 Bratislava, Slovakia
| | - Vladimira Durmanova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08 Bratislava, Slovakia
| | - Magda Suchankova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08 Bratislava, Slovakia
| | - Boris Kollarik
- Department of Urology, Saint Cyril and Methodius Hospital, Antolska 11, 851 07 Bratislava, Slovakia
| | - Patrik Hesko
- Department of Urology, Saint Cyril and Methodius Hospital, Antolska 11, 851 07 Bratislava, Slovakia
| | - Patrik Palacka
- 2nd Department of Oncology, Faculty of Medicine, Comenius University in Bratislava, Kolarska 12, 812 50 Bratislava, Slovakia
| | - Maria Bucova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08 Bratislava, Slovakia
| | - Luba Hunakova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08 Bratislava, Slovakia
| |
Collapse
|
28
|
Weng L, Hong H, Zhang Q, Xiao C, Zhang Q, Wang Q, Huang J, Lai D. Sleep Deprivation Triggers the Excessive Activation of Ovarian Primordial Follicles via β2 Adrenergic Receptor Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402393. [PMID: 39229959 PMCID: PMC11538700 DOI: 10.1002/advs.202402393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/23/2024] [Indexed: 09/05/2024]
Abstract
Sleep deprivation (SD) is observed to adversely affect the reproductive health of women. However, its precise physiological mechanisms remain largely elusive. In this study, using a mouse model of SD, it is demonstrated that SD induces the depletion of ovarian primordial follicles, a phenomenon not attributed to immune-mediated attacks or sympathetic nervous system activation. Rather, the excessive secretion of stress hormones, namely norepinephrine (NE) and epinephrine (E), by overactive adrenal glands, has emerged as a key mediator. The communication pathway mediated by the KIT ligand (KITL)-KIT between granulosa cells and oocytes plays a pivotal role in primordial follicle activation. SD heightened the levels of NE/E that stimulates the activation of the KITL-KIT/PI3K and mTOR signaling cascade in an β2 adrenergic receptor (ADRB2)-dependent manner, thereby promoting primordial follicle activation and consequent primordial follicle loss in vivo. In vitro experiments further corroborate these observations, revealing that ADRB2 upregulates KITL expression in granulosa cells via the activation of the downstream cAMP/PKA pathway. Together, these results reveal the significant involvement of ADRB2 signaling in the depletion of ovarian primordial follicles under sleep-deprived conditions. Additionally, ADRB2 antagonists are proposed for the treatment or prevention of excessive activation of primordial follicles induced by SD.
Collapse
Affiliation(s)
- Lichun Weng
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Hanqing Hong
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qinyu Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Chengqi Xiao
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qian Wang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Ju Huang
- Songjiang Hospital and Songjiang Research InstituteShanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghai201600China
| | - Dongmei Lai
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| |
Collapse
|
29
|
Reznik E, Torjani A. Mechanisms of stress-attributed breast cancer incidence and progression. Cancer Causes Control 2024; 35:1413-1432. [PMID: 39012513 DOI: 10.1007/s10552-024-01884-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 07/17/2024]
Abstract
Breast cancer is the most commonly diagnosed cancer and the second leading cause of cancer deaths in women, with psychosocial stress commonly cited by patients as one of its causes. While there is conflicting epidemiological evidence investigating the association between psychosocial stress and breast cancer incidence and progression, there is reason to believe that interventions aimed at reducing stress pharmacologically or psychologically may improve breast cancer outcomes. The aim of this review is to discuss the molecular and biological mechanisms of stress-attributed breast cancer incidence and progression, including the induction of the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS), as well as decreased immune function and stress hormone-induced resistance to chemotherapy. Moreover, these mechanisms have been cited as potential therapeutic targets of pharmacologic and psychological interventions that may improve the care, well-being and survival of breast cancer patients. Further research is recommended to investigate whether interventions in the primary care setting for women with risk factors for breast cancer development may lead to a decreased incidence of invasive breast tumors.
Collapse
Affiliation(s)
- Elizabeth Reznik
- Department of Internal Medicine, NewYork-Presbyterian Weill Cornell, New York, NY, USA.
| | - Ava Torjani
- Department of Ophthalmology, Keck School of Medicine of USC, Los Angeles, CA, USA
| |
Collapse
|
30
|
Martínez-Lira JL, Hernández-Gallegos E, DE Guadalupe Chávez-López M, Villalobos-Valencia R, Camacho J. The Effects of Nebivolol-Gefitinib-Loratadine Against Lung Cancer Cell Lines. In Vivo 2024; 38:2688-2695. [PMID: 39477390 PMCID: PMC11535926 DOI: 10.21873/invivo.13746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND/AIM Non-small-cell lung cancer (NSCLC) is the most frequently diagnosed malignancy and the first cause of cancer-related death. Thus, finding alternative therapeutic options is crucial. Drug repurposing offers therapeutic options in a simplified and affordable manner, especially to cancer patients in developing countries. Several drugs including antihistamines and beta-adrenergic receptor blockers (beta-blockers) display antiproliferative properties on cancer cells. Interestingly, NSCLC patients who had used either antihistamines or beta-blockers showed improved response to chemotherapy or reduced mortality in comparison to non-users of any of these drugs. However, combination therapy is gaining substantial interest in many cancers including non-EGFR mutated NSCLC. Here, we investigated the antineoplastic effect of the combination of the antihistamine loratadine, the beta-blocker nebivolol, and the tyrosine-kinase inhibitor gefitinib on NSCLC cell lines. MATERIALS AND METHODS A-549 and NCI-H1975 cell lines were used. The effect of nebivolol, gefitinib, and loratadine on the metabolic activity was studied using the MTT assay. The inhibitory concentrations (IC20 and IC50) were calculated and used in the drug-combination experiments. Apoptosis was investigated using flow cytometry; and cell survival using the colony formation assay. RESULTS The combination nebivolol-loratadine-gefitinib produced a significant synergistic effect on inhibiting the metabolic activity and colony formation, as well as on promoting apoptosis in both cell lines. Noteworthy, the effect on the cell line carrying the EGFR mutation (NCI-H1975) was very similar to the cell line that does not exhibit such mutation (A-549 cells). CONCLUSION The nebivolol-gefitinib-loratadine combination may be a promising alternative for lung cancer treatment.
Collapse
Affiliation(s)
- José Luis Martínez-Lira
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
- UMAE, Hospital de Especialidades Nο. 25, Instituto Mexicano del Seguro Social, Monterrey Nuevo León, México
| | - Elisabeth Hernández-Gallegos
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - María DE Guadalupe Chávez-López
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - Ricardo Villalobos-Valencia
- Departamento de Oncología Médica, Hospital de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México;
| |
Collapse
|
31
|
Othaim AA, Alasiri G, Alfahed A. Therapeutic, Clinicopathological, and Molecular Correlates of PRKACA Expression in Gastrointestinal Cancers. Pharmaceuticals (Basel) 2024; 17:1263. [PMID: 39458904 PMCID: PMC11510541 DOI: 10.3390/ph17101263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES PRKACA alterations have clear diagnostic and biological roles in the fibrolamellar variant of hepatocellular carcinoma and a potential predictive role in that cancer type. However, the roles of PRKACA have not been comprehensively examined in gastric and colorectal cancers (GC and CRC). This study, therefore, sought to investigate the roles of PRKACA expression in GC and CRC. METHODS The clinico-genomic data of 441 GC and 629 CRC cases were analyzed for therapeutic, clinicopathological, and biological correlates using appropriate bioinformatics and statistical tools. Furthermore, the deregulation of PRKACA expression in GC and CRC was investigated using correlative and regression analyses. RESULTS The results showed that PRKACA expression subsets were enriched for gene targets of chemotherapeutics, tyrosine kinase, and β-adrenergic inhibitors. Moreover, high PRKACA expression was associated with adverse clinicopathological and genomic features of GC and CRC. Gene Ontology Enrichment Analysis also showed that PRKACA-high subsets of the GI cancers were enriched for the biological and molecular functions that are associated with cell motility, invasion, and metastasis but not cell proliferation. Finally, multiple regression analyses identified multiple methylation loci, transcription factors, miRNA species, and PRKACA copy number changes that deregulated PRKACA expression in GC and CRC. CONCLUSIONS This study has identified potential predictive and clinicopathological roles for PRKACA expression in GI cancers and has added to the growing body of knowledge on the deregulation of PRKACA in cancer.
Collapse
Affiliation(s)
- Ayoub Al Othaim
- Department of Medical Laboratories, College of Applied Medical Sciences, Majmaah University, Al-Majmaah 11952, Saudi Arabia;
| | - Glowi Alasiri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia;
| | - Abdulaziz Alfahed
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| |
Collapse
|
32
|
Ma X, Deng K, Sun Y, Wu M. Research trends on cancer neuroscience: a bibliometric and visualized analysis. Front Neurosci 2024; 18:1408306. [PMID: 39268034 PMCID: PMC11390534 DOI: 10.3389/fnins.2024.1408306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Background Recently, cancer neuroscience has become the focus for scientists. Interactions between the nervous system and cancer (both systemic and local) can regulate tumorigenesis, progression, treatment resistance, compromise of anti-cancer immunity, and provocation of tumor-promoting inflammation. We assessed the related research on cancer neuroscience through bibliometric analysis and explored the research status and hotspots from 2020 to 2024. Methods Publications on cancer neuroscience retrieved from the Web of Science Core Collection. CiteSpace, VOSviewer, and Scimago Graphica were used to analyze and visualize the result. Results A total of 744 publications were retrieved, with an upward trend in the overall number of articles published over the last 5 years. As it has the highest number of publications (n = 242) and citations (average 13.63 citations per article), the United States holds an absolute voice in the field of cancer neuroscience. The most productive organizations and journals were Shanghai Jiaotong University (n = 24) and Cancers (n = 45), respectively. Monje M (H-index = 53), Hondermarck H (H-index = 42), and Amit M (H-index = 39) were the three researchers who have contributed most to the field. From a global perspective, research hotspots in cancer neuroscience comprise nerve/neuron-tumor cell interactions, crosstalk between the nervous system and other components of the tumor microenvironment (such as immune cells), as well as the impact of tumors and tumor therapies on nervous system function. Conclusion The United States and European countries are dominating the field of cancer neuroscience, while developing countries such as China are growing rapidly but with limited impact. The next focal point in this field is likely to be neurotrophic factors. Cancer neuroscience is still in its infancy, which means that many of the interactions and mechanisms between the nervous system and cancer are not yet fully understood. Further investigation is necessary to probe the interactions of the nervous system with cancer cell subpopulations and other components of the tumor microenvironment.
Collapse
Affiliation(s)
- Xinru Ma
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Kun Deng
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yingnan Sun
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Minghua Wu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| |
Collapse
|
33
|
Zhou Y, Chu P, Wang Y, Li N, Gao Q, Wang S, Wei J, Xue G, Zhao Y, Jia H, Song J, Zhang Y, Pang Y, Zhu H, Sun J, Ma S, Su C, Hu B, Zhao Z, Zhang H, Lu J, Wang J, Wang H, Sun Z, Fang D. Epinephrine promotes breast cancer metastasis through a ubiquitin-specific peptidase 22-mediated lipolysis circuit. SCIENCE ADVANCES 2024; 10:eado1533. [PMID: 39151008 PMCID: PMC11328899 DOI: 10.1126/sciadv.ado1533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/10/2024] [Indexed: 08/18/2024]
Abstract
Chronic stress-induced epinephrine (EPI) accelerates breast cancer progression and metastasis, but the molecular mechanisms remain unclear. Herein, we found a strong positive correlation between circulating EPI levels and the tumoral expression of ubiquitin-specific peptidase 22 (USP22) in patients with breast cancer. USP22 facilitated EPI-induced breast cancer progression and metastasis by enhancing adipose triglyceride lipase (ATGL)-mediated lipolysis. Targeted USP22 deletion decreased ATGL expression and lipolysis, subsequently inhibiting EPI-mediated breast cancer lung metastasis. USP22 acts as a bona fide deubiquitinase for the Atgl gene transcription factor FOXO1, and EPI architects a lipolysis signaling pathway to stabilize USP22 through AKT-mediated phosphorylation. Notably, USP22 phosphorylation levels are positively associated with EPI and with downstream pathways involving both FOXO1 and ATGL in breast cancers. Pharmacological USP22 inhibition synergized with β-blockers in treating preclinical xenograft breast cancer models. This study reveals a molecular pathway behind EPI's tumor-promoting effects and provides a strong rationale for combining USP22 inhibition with β-blockers to treat aggressive breast cancer.
Collapse
Affiliation(s)
- Yuanzhang Zhou
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Peng Chu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ya Wang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Na Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Qiong Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shengnan Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Juncheng Wei
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guoqing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Yue Zhao
- Department of Clinical Laboratory, Dalian Municipal Central Hospital, Dalian 116000, China
| | - Huijun Jia
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Jiankun Song
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Yue Zhang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Yujie Pang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Houyu Zhu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Jia Sun
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Suxian Ma
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Chen Su
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Bingjin Hu
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Zhuoyue Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Hui Zhang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Janice Lu
- Department of Medicine & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jian Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Hongjiang Wang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Zhaolin Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Deyu Fang
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
34
|
Ruggiero E, Di Castelnuovo A, Costanzo S, Esposito S, De Curtis A, Persichillo M, Cerletti C, Donati MB, de Gaetano G, Iacoviello L, Bonaccio M. Olive oil consumption is associated with lower cancer, cardiovascular and all-cause mortality among Italian adults: prospective results from the Moli-sani Study and analysis of potential biological mechanisms. Eur J Clin Nutr 2024; 78:684-693. [PMID: 38704428 DOI: 10.1038/s41430-024-01442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Olive oil consumption has been reportedly associated with lower mortality rates, mostly from cardiovascular diseases, but its potential impact on cancer death remains controversial. Moreover, biological mechanisms possibly linking olive oil consumption to mortality outcomes remain unexplored. METHODS We longitudinally analysed data on 22,892 men and women from the Moli-sani Study in Italy (follow-up 13.1 y), to examine the association of olive oil consumption with mortality. Dietary data were collected at baseline (2005-2010) through a 188-item FFQ, and olive oil consumption was standardised to a 10 g tablespoon (tbsp) size. Diet quality was assessed through a Mediterranean diet score. Multivariable-adjusted Cox proportional hazard models, also including diet quality, were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). The potential mediating role of inflammatory, metabolic, cardiovascular and renal biomarkers on the association between olive oil intake and mortality was evaluated on the basis of change-in-estimate and associated p values. RESULTS Multivariable HRs for all-cause, cancer, cardiovascular and other cause mortality associated with high (>3 tbsp/d) versus low (≤1.5 tbsp/d) olive oil consumption were 0.80 (0.69-0.94), 0.77 (0.59-0.99), 0.75 (0.58-0.97) and 0.97 (0.73-1.29), respectively. Taken together, the investigated biomarkers attenuated the association of olive oil consumption with all-cause and cancer mortality by 21.2% and 13.7%, respectively. CONCLUSIONS Higher olive oil consumption was associated with lower cancer, cardiovascular and all-cause mortality rates, independent of overall diet quality. Known risk factors for chronic diseases only in part mediated such associations suggesting that other biological pathways are potentially involved in this relationship.
Collapse
Affiliation(s)
- Emilia Ruggiero
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Augusto Di Castelnuovo
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Simona Costanzo
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Simona Esposito
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Amalia De Curtis
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Mariarosaria Persichillo
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Chiara Cerletti
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Maria Benedetta Donati
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Giovanni de Gaetano
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy.
- Department of Medicine and Surgery, LUM University "Giuseppe Degennaro", Casamassima, BA, Italy.
| | - Marialaura Bonaccio
- Department of Epidemiology and Prevention, IRCCS NEUROMED, via dell'Elettronica, 86077, Pozzilli, IS, Italy
| |
Collapse
|
35
|
Scott OW, TinTin S, Cavadino A, Elwood JM. Beta-blocker use and breast cancer outcomes: a meta-analysis. Breast Cancer Res Treat 2024; 206:443-463. [PMID: 38837086 PMCID: PMC11208256 DOI: 10.1007/s10549-024-07263-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/18/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE Beta blockers (BBs) are commonly used cardiovascular medications, and their association with breast cancer outcomes has been examined in several previous observational studies and meta-analyses. In this study, an updated meta-analysis was undertaken to ascertain the association between BBs and both breast cancer death (BCD) and breast cancer recurrence (BCR). METHODS Articles were sourced from various databases up until the 14th of August 2023. Effect estimates were pooled using the random effects model, and the Higgins I2 statistic was computed to ascertain heterogeneity. Subgroup analyses were conducted by the potential for immortal time bias (ITB), the exposure period (prediagnosis vs postdiagnosis), and type of BB (selective vs non-selective). Publication bias was assessed using funnel plots and Egger's regression tests. RESULTS Twenty-four studies were included. Pooled results showed that there was no statistically significant association between BB use and both BCD (19 studies, hazard ratio = 0.90, 95% CI 0.78-1.04) and BCR (16 studies, HR = 0.87, 95% CI 0.71-1.08). After removing studies with ITB, the associations were attenuated towards the null. There was no effect modification for either outcome when stratifying by the exposure period or type of BB. There was clear evidence of publication bias for both outcomes. CONCLUSION In this meta-analysis, we found no evidence of an association between BB use and both BCD and BCR. Removing studies with ITB attenuated the associations towards the null, but there was no effect modification by the exposure period or type of BB.
Collapse
Affiliation(s)
- Oliver William Scott
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| | - Sandar TinTin
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Alana Cavadino
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - J Mark Elwood
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
36
|
Lu J, Zhang X, Su K, Luo H, Liu C, Yang Y, He B, Wang C, Zhao Z, Liu X, Wang X, Meng P, Lv D, Wang C, Kelley KW, Wang L, Cui B, Liu Q, Peng F. Olanzapine suppresses mPFC activity-norepinephrine releasing to alleviate CLOCK-enhanced cancer stemness under chronic stress. Cell Commun Signal 2024; 22:375. [PMID: 39054537 PMCID: PMC11270788 DOI: 10.1186/s12964-024-01747-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Olanzapine (OLZ) reverses chronic stress-induced anxiety. Chronic stress promotes cancer development via abnormal neuro-endocrine activation. However, how intervention of brain-body interaction reverses chronic stress-induced tumorigenesis remains elusive. METHODS KrasLSL-G12D/WT lung cancer model and LLC1 syngeneic tumor model were used to study the effect of OLZ on cancer stemness and anxiety-like behaviors. Cancer stemness was evaluated by qPCR, western-blotting, immunohistology staining and flow-cytometry analysis of stemness markers, and cancer stem-like function was assessed by serial dilution tumorigenesis in mice and extreme limiting dilution analysis in primary tumor cells. Anxiety-like behaviors in mice were detected by elevated plus maze and open field test. Depression-like behaviors in mice were detected by tail suspension test. Anxiety and depression states in human were assessed by Hospital Anxiety and Depression Scale (HADS). Chemo-sensitivity of lung cancer was assessed by in vivo syngeneic tumor model and in vitro CCK-8 assay in lung cancer cell lines. RESULTS In this study, we found that OLZ reversed chronic stress-enhanced lung tumorigenesis in both KrasLSL-G12D/WT lung cancer model and LLC1 syngeneic tumor model. OLZ relieved anxiety and depression-like behaviors by suppressing neuro-activity in the mPFC and reducing norepinephrine (NE) releasing under chronic stress. NE activated ADRB2-cAMP-PKA-CREB pathway to promote CLOCK transcription, leading to cancer stem-like traits. As such, CLOCK-deficiency or OLZ reverses NE/chronic stress-induced gemcitabine (GEM) resistance in lung cancer. Of note, tumoral CLOCK expression is positively associated with stress status, serum NE level and poor prognosis in lung cancer patients. CONCLUSION We identify a new mechanism by which OLZ ameliorates chronic stress-enhanced tumorigenesis and chemoresistance. OLZ suppresses mPFC-NE-CLOCK axis to reverse chronic stress-induced anxiety-like behaviors and lung cancer stemness. Decreased NE-releasing prevents activation of ADRB2-cAMP-PKA-CREB pathway to inhibit CLOCK transcription, thus reversing lung cancer stem-like traits and chemoresistance under chronic stress.
Collapse
Affiliation(s)
- Jinxin Lu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- Department of Oncology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huandong Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Congcong Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yuqing Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Bin He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhuoran Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xianxian Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xu Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Peixuan Meng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Chunli Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Keith W Kelley
- Department of Pathology, College of Medicine, Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ling Wang
- Department of Oncology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
37
|
Cavalu S, Saber S, Amer AE, Hamad RS, Abdel-Reheim MA, Elmorsy EA, Abdelhamid AM. The multifaceted role of beta-blockers in overcoming cancer progression and drug resistance: Extending beyond cardiovascular disorders. FASEB J 2024; 38:e23813. [PMID: 38976162 DOI: 10.1096/fj.202400725rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/06/2024] [Accepted: 06/26/2024] [Indexed: 07/09/2024]
Abstract
Beta-blockers are commonly used medications that antagonize β-adrenoceptors, reducing sympathetic nervous system activity. Emerging evidence suggests that beta-blockers may also have anticancer effects and help overcome drug resistance in cancer treatment. This review summarizes the contribution of different isoforms of beta-adrenoceptors in cancer progression, the current preclinical and clinical data on associations between beta-blockers use and cancer outcomes, as well as their ability to enhance responses to chemotherapy and other standard therapies. We discuss proposed mechanisms, including effects on angiogenesis, metastasis, cancer stem cells, and apoptotic pathways. Overall, results from epidemiological studies and small clinical trials largely indicate the beneficial effects of beta-blockers on cancer progression and drug resistance. However, larger randomized controlled trials are needed to firmly establish their clinical efficacy and optimal utilization as adjuvant agents in cancer therapy.
Collapse
Affiliation(s)
- Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Ahmed E Amer
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amir Mohamed Abdelhamid
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
38
|
Uher O, Hadrava Vanova K, Taïeb D, Calsina B, Robledo M, Clifton-Bligh R, Pacak K. The Immune Landscape of Pheochromocytoma and Paraganglioma: Current Advances and Perspectives. Endocr Rev 2024; 45:521-552. [PMID: 38377172 PMCID: PMC11244254 DOI: 10.1210/endrev/bnae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors derived from neural crest cells from adrenal medullary chromaffin tissues and extra-adrenal paraganglia, respectively. Although the current treatment for PPGLs is surgery, optimal treatment options for advanced and metastatic cases have been limited. Hence, understanding the role of the immune system in PPGL tumorigenesis can provide essential knowledge for the development of better therapeutic and tumor management strategies, especially for those with advanced and metastatic PPGLs. The first part of this review outlines the fundamental principles of the immune system and tumor microenvironment, and their role in cancer immunoediting, particularly emphasizing PPGLs. We focus on how the unique pathophysiology of PPGLs, such as their high molecular, biochemical, and imaging heterogeneity and production of several oncometabolites, creates a tumor-specific microenvironment and immunologically "cold" tumors. Thereafter, we discuss recently published studies related to the reclustering of PPGLs based on their immune signature. The second part of this review discusses future perspectives in PPGL management, including immunodiagnostic and promising immunotherapeutic approaches for converting "cold" tumors into immunologically active or "hot" tumors known for their better immunotherapy response and patient outcomes. Special emphasis is placed on potent immune-related imaging strategies and immune signatures that could be used for the reclassification, prognostication, and management of these tumors to improve patient care and prognosis. Furthermore, we introduce currently available immunotherapies and their possible combinations with other available therapies as an emerging treatment for PPGLs that targets hostile tumor environments.
Collapse
Affiliation(s)
- Ondrej Uher
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - Katerina Hadrava Vanova
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - David Taïeb
- Department of Nuclear Medicine, CHU de La Timone, Marseille 13005, France
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Familiar Cancer Clinical Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid 28029, Spain
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, Sydney 2065, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney 2065, NSW, Australia
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| |
Collapse
|
39
|
Gudenkauf LM, Hathaway CA, Carroll JE, Small BJ, Li X, Hoogland AI, Castro E, Armaiz-Pena GN, Oswald LB, Jim HS, Tworoger SS, Gonzalez BD. Inequities in the Impacts of Hurricanes and Other Extreme Weather Events for Cancer Survivors. Cancer Epidemiol Biomarkers Prev 2024; 33:771-778. [PMID: 38385842 PMCID: PMC11147728 DOI: 10.1158/1055-9965.epi-23-1029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/12/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
In this minireview, we examine the impacts of hurricanes and other extreme weather events on cancer survivors, focusing on structural and social determinants of health. We briefly explore influences on biological, psychosocial, and behavioral outcomes and discuss risk and resilience factors in cancer survivorship during and after hurricanes. Our goal is to inform future directions for research that can identify areas in which we can most efficiently improve cancer outcomes and inform changes in health systems, clinical practice, and public health policies. This timely minireview provides researchers and clinicians with an overview of challenges and opportunities for improving disaster preparedness and response for cancer survivors.
Collapse
Affiliation(s)
- Lisa M. Gudenkauf
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Judith E. Carroll
- Department of Psychiatry and Behavioral Sciences, University of California, Los Angeles, CA, United States
| | - Brent J. Small
- School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xiaoyin Li
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States
| | - Aasha I. Hoogland
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States
| | - Eida Castro
- School of Behavior and Brain Sciences, Ponce Health Sciences University, Ponce, PR, United States
| | - Guillermo N. Armaiz-Pena
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States
| | - Laura B. Oswald
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States
| | - Heather S.L. Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States
| | - Shelley S. Tworoger
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, United States
| | - Brian D. Gonzalez
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
40
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
41
|
Gonzalez-Llerena JL, Espinosa-Rodriguez BA, Treviño-Almaguer D, Mendez-Lopez LF, Carranza-Rosales P, Gonzalez-Barranco P, Guzman-Delgado NE, Romo-Mancillas A, Balderas-Renteria I. Cordycepin Triphosphate as a Potential Modulator of Cellular Plasticity in Cancer via cAMP-Dependent Pathways: An In Silico Approach. Int J Mol Sci 2024; 25:5692. [PMID: 38891880 PMCID: PMC11171877 DOI: 10.3390/ijms25115692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Cordycepin, or 3'-deoxyadenosine, is an adenosine analog with a broad spectrum of biological activity. The key structural difference between cordycepin and adenosine lies in the absence of a hydroxyl group at the 3' position of the ribose ring. Upon administration, cordycepin can undergo an enzymatic transformation in specific tissues, forming cordycepin triphosphate. In this study, we conducted a comprehensive analysis of the structural features of cordycepin and its derivatives, contrasting them with endogenous purine-based metabolites using chemoinformatics and bioinformatics tools in addition to molecular dynamics simulations. We tested the hypothesis that cordycepin triphosphate could bind to the active site of the adenylate cyclase enzyme. The outcomes of our molecular dynamics simulations revealed scores that are comparable to, and superior to, those of adenosine triphosphate (ATP), the endogenous ligand. This interaction could reduce the production of cyclic adenosine monophosphate (cAMP) by acting as a pseudo-ATP that lacks a hydroxyl group at the 3' position, essential to carry out nucleotide cyclization. We discuss the implications in the context of the plasticity of cancer and other cells within the tumor microenvironment, such as cancer-associated fibroblast, endothelial, and immune cells. This interaction could awaken antitumor immunity by preventing phenotypic changes in the immune cells driven by sustained cAMP signaling. The last could be an unreported molecular mechanism that helps to explain more details about cordycepin's mechanism of action.
Collapse
Affiliation(s)
- Jose Luis Gonzalez-Llerena
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
- Center for Research on Nutrition and Public Health, School of Public Health and Nutrition, Autonomous University of Nuevo Leon, Monterrey 66460, Mexico;
| | - Bryan Alejandro Espinosa-Rodriguez
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Daniela Treviño-Almaguer
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Luis Fernando Mendez-Lopez
- Center for Research on Nutrition and Public Health, School of Public Health and Nutrition, Autonomous University of Nuevo Leon, Monterrey 66460, Mexico;
| | - Pilar Carranza-Rosales
- Laboratory of Cell Biology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Mexico;
| | - Patricia Gonzalez-Barranco
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Nancy Elena Guzman-Delgado
- Health Research Division, High Specialty Medical Unit, Cardiology Hospital N. 34. Mexican Social Security Institute, Monterrey 64360, Mexico;
| | - Antonio Romo-Mancillas
- Computer Aided Drug Design and Synthesis Group, School of Chemistry, Autonomous University of Queretaro, Queretaro 76010, Mexico
| | - Isaias Balderas-Renteria
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| |
Collapse
|
42
|
Sayour NV, Paál ÁM, Ameri P, Meijers WC, Minotti G, Andreadou I, Lombardo A, Camilli M, Drexel H, Grove EL, Dan GA, Ivanescu A, Semb AG, Savarese G, Dobrev D, Crea F, Kaski JC, de Boer RA, Ferdinandy P, Varga ZV. Heart failure pharmacotherapy and cancer: pathways and pre-clinical/clinical evidence. Eur Heart J 2024; 45:1224-1240. [PMID: 38441940 PMCID: PMC11023004 DOI: 10.1093/eurheartj/ehae105] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 04/08/2024] Open
Abstract
Heart failure (HF) patients have a significantly higher risk of new-onset cancer and cancer-associated mortality, compared to subjects free of HF. While both the prevention and treatment of new-onset HF in patients with cancer have been investigated extensively, less is known about the prevention and treatment of new-onset cancer in patients with HF, and whether and how guideline-directed medical therapy (GDMT) for HF should be modified when cancer is diagnosed in HF patients. The purpose of this review is to elaborate and discuss the effects of pillar HF pharmacotherapies, as well as digoxin and diuretics on cancer, and to identify areas for further research and novel therapeutic strategies. To this end, in this review, (i) proposed effects and mechanisms of action of guideline-directed HF drugs on cancer derived from pre-clinical data will be described, (ii) the evidence from both observational studies and randomized controlled trials on the effects of guideline-directed medical therapy on cancer incidence and cancer-related outcomes, as synthetized by meta-analyses will be reviewed, and (iii) considerations for future pre-clinical and clinical investigations will be provided.
Collapse
Affiliation(s)
- Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Ágnes M Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Italian IRCCS Cardiology Network, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Wouter C Meijers
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Giorgio Minotti
- University Campus Bio-Medico, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Ioanna Andreadou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation & Treatment (VIVIT), Carinagasse 47, A-6800 Feldkirch, Austria
| | - Erik Lerkevang Grove
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Gheorghe Andrei Dan
- Carol Davila University of Medicine and Pharmacy, Colentina University Hospital, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Andreea Ivanescu
- Carol Davila University of Medicine and Pharmacy, Colentina University Hospital, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Anne Grete Semb
- Division of Research and Innovation, REMEDY-Centre for Treatment of Rheumatic and Musculoskeletal Diseases, Diakonhjemmet Hospital, Oslo, Norway
| | - Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular and Neuro Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, QC, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Juan-Carlos Kaski
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Rudolf A de Boer
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- Pharmahungary Group, Szeged, Hungary
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
| |
Collapse
|
43
|
Dumas E, Grandal Rejo B, Gougis P, Houzard S, Abécassis J, Jochum F, Marande B, Ballesta A, Del Nery E, Dubois T, Alsafadi S, Asselain B, Latouche A, Espie M, Laas E, Coussy F, Bouchez C, Pierga JY, Le Bihan-Benjamin C, Bousquet PJ, Hotton J, Azencott CA, Reyal F, Hamy AS. Concomitant medication, comorbidity and survival in patients with breast cancer. Nat Commun 2024; 15:2966. [PMID: 38580683 PMCID: PMC10997660 DOI: 10.1038/s41467-024-47002-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/14/2024] [Indexed: 04/07/2024] Open
Abstract
Between 30% and 70% of patients with breast cancer have pre-existing chronic conditions, and more than half are on long-term non-cancer medication at the time of diagnosis. Preliminary epidemiological evidence suggests that some non-cancer medications may affect breast cancer risk, recurrence, and survival. In this nationwide cohort study, we assessed the association between medication use at breast cancer diagnosis and survival. We included 235,368 French women with newly diagnosed non-metastatic breast cancer. In analyzes of 288 medications, we identified eight medications positively associated with either overall survival or disease-free survival: rabeprazole, alverine, atenolol, simvastatin, rosuvastatin, estriol (vaginal or transmucosal), nomegestrol, and hypromellose; and eight medications negatively associated with overall survival or disease-free survival: ferrous fumarate, prednisolone, carbimazole, pristinamycin, oxazepam, alprazolam, hydroxyzine, and mianserin. Full results are available online from an interactive platform ( https://adrenaline.curie.fr ). This resource provides hypotheses for drugs that may naturally influence breast cancer evolution.
Collapse
Affiliation(s)
- Elise Dumas
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France
- INSERM, U900, 75005, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, 75006, Paris, France
| | - Beatriz Grandal Rejo
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France
| | - Paul Gougis
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France
| | - Sophie Houzard
- Health Data and Assessment, Health Survey Data Science and Assessment Division, French National Cancer Institute (Institut National du Cancer INCa), 92100, Boulogne-Billancourt, France
| | - Judith Abécassis
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France
- INRIA, Paris-Saclay University, CEA, Palaiseau, 91120, France
| | - Floriane Jochum
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France
- Department of Gynecology, Strasbourg University Hospital, Strasbourg, France
| | - Benjamin Marande
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France
| | - Annabelle Ballesta
- INSERM UMR-S 900, Institut Curie, MINES ParisTech CBIO, PSL Research University, 92210, Saint-Cloud, France
| | - Elaine Del Nery
- Département de Recherche Translationnelle - Plateforme Biophenics, PICT-IBISA, PSL Research University, Paris, France
| | - Thierry Dubois
- Institut Curie - PSL Research University Translational Research Department Breast Cancer Biology Group 26 rue d'Ulm, 75005, Paris, France
| | - Samar Alsafadi
- Institut Curie, PSL Research University, Uveal Melanoma Group, Translational Research Department, Paris, France
| | | | - Aurélien Latouche
- INSERM, U900, 75005, Paris, France
- INSERM UMR-S 900, Institut Curie, MINES ParisTech CBIO, PSL Research University, 92210, Saint-Cloud, France
- Conservatoire National des Arts et Métiers, Paris, France
| | - Marc Espie
- Breast diseases Center Hôpital saint Louis APHP, Université Paris Cité, Paris, France
| | - Enora Laas
- Department of Surgical Oncology, Université Paris Cité, Institut Curie, 75005, Paris, France
| | - Florence Coussy
- Department of Medical Oncology, Université Paris Cité, Institut Curie, 75005, Paris, France
| | - Clémentine Bouchez
- Breast diseases Center Hôpital saint Louis APHP, Université Paris Cité, Paris, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Université Paris Cité, Institut Curie, 75005, Paris, France
| | - Christine Le Bihan-Benjamin
- Health Data and Assessment, Health Survey Data Science and Assessment Division, French National Cancer Institute (Institut National du Cancer INCa), 92100, Boulogne-Billancourt, France
| | - Philippe-Jean Bousquet
- Aix Marseille Univ, Inserm, IRD, SESSTIM, Équipe Labellisée Ligue Contre le Cancer, 13005, Marseille, France
- Health Survey Data Science and Assessment Division, French National Cancer Institute (Institut National du Cancer INCa), 92100, Boulogne-Billancourt, France
| | | | - Chloé-Agathe Azencott
- INSERM, U900, 75005, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, 75006, Paris, France
- Institut Curie, PSL Research University, Paris, France
| | - Fabien Reyal
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France.
- Department of Surgical Oncology, Université Paris Cité, Institut Curie, 75005, Paris, France.
- Department of Surgery, Institut Jean Godinot, Reims, France.
| | - Anne-Sophie Hamy
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Université Paris Cité, F-75005, Paris, France
- Department of Medical Oncology, Université Paris Cité, Institut Curie, 75005, Paris, France
| |
Collapse
|
44
|
Abstract
Although there is little direct evidence supporting that stress affects cancer incidence, it does influence the evolution, dissemination and therapeutic outcomes of neoplasia, as shown in human epidemiological analyses and mouse models. The experience of and response to physiological and psychological stressors can trigger neurological and endocrine alterations, which subsequently influence malignant (stem) cells, stromal cells and immune cells in the tumour microenvironment, as well as systemic factors in the tumour macroenvironment. Importantly, stress-induced neuroendocrine changes that can regulate immune responses have been gradually uncovered. Numerous stress-associated immunomodulatory molecules (SAIMs) can reshape natural or therapy-induced antitumour responses by engaging their corresponding receptors on immune cells. Moreover, stress can cause systemic or local metabolic reprogramming and change the composition of the gastrointestinal microbiota which can indirectly modulate antitumour immunity. Here, we explore the complex circuitries that link stress to perturbations in the cancer-immune dialogue and their implications for therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Yuting Ma
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.
| | - Guido Kroemer
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
45
|
Everatt R, Kuzmickienė I, Brasiūnienė B, Vincerževskienė I, Intaitė B, Cicėnas S, Lisauskienė I. Evaluation of antihypertensive medications use and survival in patients with ovarian cancer: a population-based retrospective cohort study. BMC Womens Health 2024; 24:155. [PMID: 38439058 PMCID: PMC10913626 DOI: 10.1186/s12905-024-02983-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Despite declining mortality in most countries and in Lithuania, ovarian cancer burden has remained high. Studies have indicated that antihypertensive medications use may help to improve ovarian cancer survival, however findings remain controversial. The aim of the study was to analyse the association between post-diagnosis antihypertensive medications intake and cancer-specific survival in ovarian cancer patients. METHODS This retrospective cohort study included 588 ovarian cancer cases diagnosed between 2013 and 2015. Hazard ratios (HR) and corresponding 95% confidence intervals (95%CI) were estimated using multivariable Cox proportional hazards models to assess associations between antihypertensive medications and ovarian cancer-specific mortality. RESULTS In total, 279 (47%) patients died during the follow-up; 242 (87%) of them died due to ovarian cancer. The risk of ovarian cancer death was reduced in angiotensin-converting enzyme inhibitors (ACE inhibitors) users vs. non-users (HR 0.55, 95% CI: 0.36-0.83). Subgroup analysis showed better ovarian cancer survival in higher dose ACE inhibitors users (HR 0.46, 95% CI: 0.28-0.77, p for trend 0.002); the effect was also stronger in age 51-65 years, stage I-III, surgery or chemotherapy treatment, pre-diagnosis ACE inhibitor users' and pre-diagnosis hypertension subgroups. The risk of cancer-specific death was slightly lower among calcium-channel blocker and angiotensin-receptor blocker users and higher among beta-blocker users as compared to non-users, however chance and confounding could not be ruled out. We found no association between the use of centrally and peripherally acting antiadrenergic agents and diuretics and risk of ovarian cancer-specific mortality. CONCLUSIONS Our findings imply that post-diagnosis use of ACE inhibitors may be associated with reduced ovarian cancer-specific mortality; however, further research is needed for the comprehensive assessment.
Collapse
Affiliation(s)
- Rūta Everatt
- Laboratory of Cancer Epidemiology, National Cancer Institute, Baublio 3B, Vilnius, LT-08406, Lithuania.
| | - Irena Kuzmickienė
- Laboratory of Cancer Epidemiology, National Cancer Institute, Baublio 3B, Vilnius, LT-08406, Lithuania
| | - Birutė Brasiūnienė
- Department of Medical Oncology, National Cancer Institute, Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Birutė Intaitė
- Department of Gynaecologic Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Saulius Cicėnas
- Department of Thoracic Surgery and Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Ingrida Lisauskienė
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
46
|
Imran S, Rao MS, Shah MH, Gaur A, Guernaoui AE, Roy S, Roy S, Bharadwaj HR, Awuah WA. Evolving perspectives in reverse cardio-oncology: A review of current status, pathophysiological insights, and future directives. Curr Probl Cardiol 2024; 49:102389. [PMID: 38184129 DOI: 10.1016/j.cpcardiol.2024.102389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/08/2024]
Abstract
Cardiovascular disease (CVD) and cancer are leading causes of mortality worldwide, traditionally linked through adverse effects of cancer therapies on cardiovascular health. However, reverse cardio-oncology, a burgeoning field, shifts this perspective to examine how cardiovascular diseases influence the onset and progression of cancer. This novel approach has revealed a higher likelihood of cancer development in patients with pre-existing cardiovascular conditions, attributed to shared risk factors such as obesity, a sedentary lifestyle, and smoking. Underlying mechanisms like chronic inflammation and clonal hematopoiesis further illuminate the connections between cardiovascular ailments and cancer. This comprehensive narrative review, spanning a broad spectrum of studies, outlines the syndromic classification of cardio-oncology, the intersection of cardiovascular risk factors and oncogenesis, and the bidirectional dynamics between CVD and cancer. Additionally, the review also discusses the pathophysiological mechanisms underpinning this interconnection, examining the roles of cardiokines, genetic factors, and the effects of cardiovascular therapies and biomarkers in cancer diagnostics. Lastly, it aims to underline future directives, emphasising the need for integrated healthcare strategies, interdisciplinary research, and comprehensive treatment protocols.
Collapse
Affiliation(s)
- Shahzeb Imran
- School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Medha Sridhar Rao
- School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Muhammad Hamza Shah
- School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom; Centre for Anatomy, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Aditya Gaur
- School of Medicine, University of Central Lancashire, Preston, United Kingdom
| | - Abderrahmane El Guernaoui
- School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Subham Roy
- Hull York Medical School, University of York, York, United Kingdom
| | - Sakshi Roy
- School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | | | | |
Collapse
|
47
|
Ali M, Wani SUD, Dey T, Sridhar SB, Qadrie ZL. A common molecular and cellular pathway in developing Alzheimer and cancer. Biochem Biophys Rep 2024; 37:101625. [PMID: 38225990 PMCID: PMC10788207 DOI: 10.1016/j.bbrep.2023.101625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024] Open
Abstract
Globally cancer and Alzheimer's disease (AD) are two major diseases and still, there is no clearly defined molecular mechanism. There is an opposite relation between cancer and AD which are the proportion of emerging cancer was importantly slower in AD patients, whereas slow emerging AD in patients with cancer. In cancer, regulation of cell mechanisms is interrupted by an increase in cell survival and proliferation, while on the contrary, AD is related to augmented neuronal death, that may be either produced by or associated with amyloid-β (Aβ) and tau deposition. Stated that the probability that disruption of mechanisms takes part in the regulation of cell survival/death and might be implicated in both diseases. The mechanism of actions such as DNA-methylation, genetic polymorphisms, or another mechanism of actions that induce alteration in the action of drugs with significant roles in resolving the finding to repair and live or die might take part in the pathogenesis of these two ailments. The functions of miRNA, p53, Pin1, the Wnt signaling pathway, PI3 KINASE/Akt/mTOR signaling pathway GRK2 signaling pathway, and the pathophysiological role of oxidative stress are presented in this review as potential candidates which hypothetically describe inverse relations between cancer and AD. Innovative materials almost mutual mechanisms in the aetiology of cancer and AD advocates novel treatment approaches. Among these treatment strategies, the most promising use treatment such as tyrosine kinase inhibitor, nilotinib, protein kinase C, and bexarotene.
Collapse
Affiliation(s)
- Mohammad Ali
- Department of Pharmacology, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B.G Nagar, Nagamagala, Bellur, Karnataka, 571418, India
- Department of Pharmacy Practice, East Point College of Pharmacy, Bangalore, 560049, India
| | - Shahid Ud Din Wani
- Division of Pharmaceutics, Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, 190006, India
| | - Tathagata Dey
- Department of Pharmaceutical Chemistry, East Point College of Pharmacy, Bangalore, 560049, India
| | - Sathvik B. Sridhar
- Department of Clinical Pharmacy and Pharmacology, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, PO Box 11172, United Arab Emirates
| | | |
Collapse
|
48
|
Kolb T, Müller S, Möller P, Barth TFE, Marienfeld R. Molecular heterogeneity in histomorphologic subtypes of lung adeno carcinoma represents a challenge for treatment decision. Neoplasia 2024; 49:100955. [PMID: 38310709 PMCID: PMC10848034 DOI: 10.1016/j.neo.2023.100955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/13/2023] [Indexed: 02/06/2024]
Abstract
Lung cancer is the leading cause in cancer related death, with non-small cell lung cancer (NSCLC) being the most frequent subtype. The importance of NSCLC is reflected by the various targeted therapy options especially for NSCLC adenocarcinomas (lung adeno carcinoma (LUAD)) as well as a set of options for immune therapies. However, despite these therapy advances, the majority of patients do not show a long-term response to either targeted therapy or immune checkpoint inhibition. One reason for treatment failure appears to be the NSCLC tumor heterogeneity. NSCLC heterogeneity might lead to an insufficient molecular characterization of a given sample due to the limited tumor material used for pathological assessment as the majority of analyses is performed on small biopsies. To get a more detailed insight into the tumor heterogeneity of NSCLC LUAD, especially in the light of its different histomorphological growth patterns, we analysed isolated NSCLC growth pattern areas and the corresponding entire tumor samples of a cohort of 31 NSLCS LUAD patients and compared their mutational landscape and their expression profiles. While significant differences of complex biomarkers, like tumor mutational burden (TMB) or microsatellite instability (MSI), were not detected between the five growth patterns -lepidic, papillary, micropapillary, acinar, and solid- we observed various subclonal mutations and copy number variants. Moreover, RNASeq analysis revealed growth pattern specific expression profiles affecting cellular processes like apoptosis, metastasis and proliferation. Taken together, our data provide novel insights into the tumor heterogeneity of LUAD required to overcome tumor heterogeneity related therapy resistance.
Collapse
Affiliation(s)
- Tobias Kolb
- Institute of Pathology, Ulm University, Ulm, Germany
| | - Sarah Müller
- Institute of Pathology, Ulm University, Ulm, Germany
| | - Peter Möller
- Institute of Pathology, Ulm University, Ulm, Germany
| | | | | |
Collapse
|
49
|
Sykes DJ, Solanki S, Chukkapalli S, Williams K, Newman EA, Resnicow K, Shah YM. Structural enrichment attenuates colitis-associated colon cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580099. [PMID: 38405737 PMCID: PMC10888747 DOI: 10.1101/2024.02.13.580099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Colorectal cancer (CRC) is a major public health concern and disproportionately impacts racial/ethnic minority populations in the US. Animal models are helpful in examining human health disparities because many stress-induced human health conditions can be recapitulated using mouse models. Azoxymethane (AOM)/ dextran sodium sulfate (DSS) treatment can be used to model colitis-associated cancers. While colitis-associated cancers account for only 2% of colon cancers, the AOM/DSS model is useful for examining links between inflammation, immunity, and colon cancer. Mice were housed in enriched and impoverished environments for 1-month prior to behavioral testing. Following behavioral testing the mice were subjected to the AOM/DSS model. While our analysis revealed no significant behavioral variances between the impoverished and enriched housing conditions, we found significant effects in tumorigenesis. Enriched mice had fewer tumors and smaller tumor volumes compared to impoverished mice. African Americans are at higher risk for early onset colorectal cancers in part due to social economic status. Furthermore, housing conditions and environment may reflect social economic status. Research aimed at understanding links between social economic status and colorectal cancer progression is important for eliminating disparities in health outcomes.
Collapse
|
50
|
Leshem Y, Etan T, Dolev Y, Nikolaevski-Berlin A, Miodovnik M, Shamai S, Merimsky O, Wolf I, Havakuk O, Tzuberi M, Topilsky Y, Banai S, Rozenbaum Z, Laufer-Perl M. The prognostic value of beta-1 blockers in patients with non-small-cell lung carcinoma treated with pembrolizumab. Int J Cardiol 2024; 397:131642. [PMID: 38065325 DOI: 10.1016/j.ijcard.2023.131642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) such as pembrolizumab have revolutionized the treatment of metastatic non-small cell lung cancer (mNSCLC). Beta-adrenergic activation contributes to cancer initiation and progression. While non-selective beta-blocker were found to improve the efficacy of ICIs therapy, the role of beta-1 (β1)-selective -blocker (β1B) in lung cancer patients is unknown. OBJECTIVE To evaluate the effect of β1B on overall survival (OS) and progression-free survival (PFS) in patients diagnosed with mNSCLC and treated with pembrolizumab. METHODS We performed a retrospective analysis of patients diagnosed with mNSCLC and treated with first-line pembrolizumab at our center. RESULTS Of 200 eligible patients, 53 (27%) were pretreated with β1B. Patients in the β1B cohort were older (73 ± 8 vs. 67 ± 10 years, p < 0.001) with a higher prevalence of cardiac risk factors and cardiovascular (CV) diseases including ischemic heart disease (32% vs. 16%, p = 0.010), heart failure (9% vs. 3%, p = 0.043) and atrial fibrillation (23% vs. 3%, p < 0.001). Compared to the non-β1B group, patient pretreated with β1B had a significant shorter median OS (12 vs. 24 months, p = 0.004) and PFS (6 vs. 8 months, p < 0.001). In a multivariate analysis, including all CV risk factors and diseases, the use of baseline β1B was a strong and independent predictor for accelerated disease progression (HR 1.92, 95%CI 1.32-2.79, p < 0.001) and shorter OS (HR 1.8, 95%, CI 1.18-2.75, p = 0.007). CONCLUSIONS The use of baseline β1B showed a strong and independent association for shorter OS and PFS in patients diagnosed with mNSCLC and treated with pembrolizumab.
Collapse
Affiliation(s)
- Yasmin Leshem
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel; Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Etan
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yardenna Dolev
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Mor Miodovnik
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sivan Shamai
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ofer Merimsky
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ido Wolf
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Havakuk
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maor Tzuberi
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yan Topilsky
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shmuel Banai
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zach Rozenbaum
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Tulane University, New Orleans, LA, United States of America
| | - Michal Laufer-Perl
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|