1
|
Kessler L, Koo C, Richter CP, Tan X. Hearing loss during chemotherapy: prevalence, mechanisms, and protection. Am J Cancer Res 2024; 14:4597-4632. [PMID: 39417180 PMCID: PMC11477841 DOI: 10.62347/okgq4382] [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: 05/15/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Ototoxicity is an often-underestimated sequela for cancer patients undergoing chemotherapy, with an incidence rate exceeding 50%, affecting approximately 4 million individuals worldwide each year. Despite the nearly 2,000 publications on chemotherapy-related ototoxicity in the past decade, the understanding of its prevalence, mechanisms, and preventative or therapeutic measures remains ambiguous and subject to debate. To date, only one drug, sodium thiosulfate, has gained FDA approval for treating ototoxicity in chemotherapy. However, its utilization is restricted. This review aims to offer clinicians and researchers a comprehensive perspective by thoroughly and carefully reviewing available data and current evidence. Chemotherapy-induced ototoxicity is characterized by four primary symptoms: hearing loss, tinnitus, vertigo, and dizziness, originating from both auditory and vestibular systems. Hearing loss is the predominant symptom. Amongst over 700 chemotherapeutic agents documented in various databases, only seven are reported to induce hearing loss. While the molecular mechanisms of the hearing loss caused by the two platinum-based drugs are extensively explored, the pathways behind the action of the other five drugs are primarily speculative, rooted in their therapeutic properties and side effects. Cisplatin attracts the majority of attention among these drugs, encompassing around two-thirds of the literature regarding ototoxicity in chemotherapy. Cisplatin ototoxicity chiefly manifests through the loss of outer hair cells, possibly resulting from damages directly by cisplatin uptake or secondary effects on the stria vascularis. Both direct and indirect influences contribute to cisplatin ototoxicity, while it is still debated which path is dominant or where the primary target of cisplatin is located. Candidates for hearing protection against cisplatin ototoxicity are also discussed, with novel strategies and methods showing promise on the horizon.
Collapse
Affiliation(s)
- Lexie Kessler
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Chail Koo
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Claus-Peter Richter
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Biomedical Engineering, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Communication Sciences and Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| | - Xiaodong Tan
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| |
Collapse
|
2
|
Dominiak K, Gostyńska A, Szulc M, Stawny M. The Anticancer Application of Delivery Systems for Honokiol and Magnolol. Cancers (Basel) 2024; 16:2257. [PMID: 38927963 PMCID: PMC11201421 DOI: 10.3390/cancers16122257] [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: 05/14/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer is a leading cause of death worldwide, and the effectiveness of treatment is consistently not at a satisfactory level. This review thoroughly examines the present knowledge and perspectives of honokiol (HON) in cancer therapeutics. The paper synthesizes critical insights into the molecular mechanisms underlying the observed anticancer effects, emphasizing both in vitro and in vivo studies. The effects of HON application, primarily in the common types of cancers, are presented. Because the therapeutic potential of HON may be limited by its physicochemical properties, appropriate delivery systems are sought to overcome this problem. This review discusses the effect of different nanotechnology-based delivery systems on the efficiency of HON. The data presented show that HON exhibits anticancer effects and can be successfully administered to the site of action. Honokiol exerts its anticancer activity through several mechanisms. Moreover, some authors used the combinations of classical anticancer drugs with HON. Such an approach is very interesting and worth further investigation. Understanding HON's multiple molecular mechanisms would provide valuable insights into how HON might be developed as an effective therapeutic. Therefore, further research is needed to explore its specific applications and optimize its efficacy in diverse cancer types.
Collapse
Affiliation(s)
- Katarzyna Dominiak
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland;
| | - Aleksandra Gostyńska
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland;
| | - Michał Szulc
- Department of Pharmacology, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland;
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland;
| |
Collapse
|
3
|
Prasher P, Fatima R, Sharma M, Tynybekov B, Alshahrani AM, Ateşşahin DA, Sharifi-Rad J, Calina D. Honokiol and its analogues as anticancer compounds: Current mechanistic insights and structure-activity relationship. Chem Biol Interact 2023; 386:110747. [PMID: 37816447 DOI: 10.1016/j.cbi.2023.110747] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/22/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023]
Abstract
Lignans are plant-derived polyphenolic compounds with a plethora of biological applications. Also, regarded as phytoestrogens, the lignans offer a variety of health benefits of which the anti-cancer effects are the most attractive. Honokiol is a lignan isolated from various parts of trees belonging to the genus Magnolia. The bioactivity of honokiol is attributed to its characteristic physical properties, which include small size and the presence of two phenolic groups that may interact with proteins in cell membranes via hydrophobic interactions, aromatic pi orbital co-valency, and hydrogen bonding. The hydrophobicity of honokiol enables its rapid dissolution in lipids and the crossing of physiological barriers, including the blood-brain barrier and cerebrospinal fluid. These factors contribute towards the high bioavailability of honokiol which further support its candidature in medicinal research. Therefore, the anticancer properties of honokiol are of particular interest as many of the contemporary anticancer drugs suffer from bioavailability drawbacks, which necessitates the identification and development of novel candidate molecules directed as anticancer chemotherapeutics. The antioncogenic profile of honokiol also arises from the regulation of various signalling pathways associated with oncogenesis, arresting of the cell cycle by regulation of cyclic proteins, upregulation of epithelial markers and downregulation of mesenchymal markers leading to the inhibition of epithelial-mesenchymal transition, and preventing the metastasis by restricting cell migration and invasion due to the downregulation of matrix-metalloproteinases. In this review, we discuss the anticancer properties of honokiol.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India.
| | - Rabab Fatima
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India.
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Arcadia Grant, Dehradun, 248007, India.
| | - Bekzat Tynybekov
- Al-Farabi Kazakh National University, Department of Biodiversity and Bioresources, Almaty, Kazakhstan.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Dilek Arslan Ateşşahin
- Fırat University, Baskil Vocational School, Department of Plant and Animal Production, 23100, Elazıg, Turkey.
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
4
|
Yang J, Shang J, Yang L, Wei D, Wang X, Deng Q, Zhong Z, Ye Y, Zhou M. Nanotechnology-Based Drug Delivery Systems for Honokiol: Enhancing Therapeutic Potential and Overcoming Limitations. Int J Nanomedicine 2023; 18:6639-6665. [PMID: 38026538 PMCID: PMC10656744 DOI: 10.2147/ijn.s431409] [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: 07/27/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Honokiol (HNK) is a small-molecule polyphenol that has garnered considerable attention due to its diverse pharmacological properties, including antitumor, anti-inflammatory, anti-bacterial, and anti-obesity effects. However, its clinical application is restricted by challenges such as low solubility, poor bioavailability, and rapid metabolism. To overcome these limitations, researchers have developed a variety of nano-formulations for HNK delivery. These nano-formulations offer advantages such as enhanced solubility, improved bioavailability, extended circulation time, and targeted drug delivery. However, existing reviews of HNK primarily focus on its clinical and pharmacological features, leaving a gap in the comprehensive evaluation of HNK delivery systems based on nanotechnology. This paper aims to bridge this gap by comprehensively reviewing different types of nanomaterials used for HNK delivery over the past 15 years. These materials encompass vesicle delivery systems, nanoparticles, polymer micelles, nanogels, and various other nanocarriers. The paper details various HNK nano-delivery strategies and summarizes their latest applications, development prospects, and future challenges. To compile this review, we conducted an extensive search using keywords such as "honokiol", "nanotechnology", and "drug delivery system" on reputable databases, including PubMed, Scopus, and Web of Science, covering the period from 2008 to 2023. Through this search, we identified and selected approximately 90 articles that met our specific criteria.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Jinlu Shang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Liuxuan Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Daiqing Wei
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Xia Wang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Qinmin Deng
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yun Ye
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
5
|
Rybak LP, Alberts I, Patel S, Al Aameri RFH, Ramkumar V. Effects of natural products on cisplatin ototoxicity and chemotherapeutic efficacy. Expert Opin Drug Metab Toxicol 2023; 19:635-652. [PMID: 37728555 DOI: 10.1080/17425255.2023.2260737] [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: 03/30/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023]
Abstract
INTRODUCTION Cisplatin is a very effective chemotherapeutic agent against a variety of solid tumors. Unfortunately, cisplatin causes permanent sensorineural hearing loss in at least two-thirds of patients treated. There are no FDA approved drugs to prevent this serious side effect. AREAS COVERED This paper reviews various natural products that ameliorate cisplatin ototoxicity. These compounds are strong antioxidants and anti-inflammatory agents. This review includes mostly preclinical studies but also discusses a few small clinical trials with natural products to minimize hearing loss from cisplatin chemotherapy in patients. The interactions of natural products with cisplatin in tumor-bearing animal models are highlighted. A number of natural products did not interfere with cisplatin anti-tumor efficacy and some agents actually potentiated cisplatin anti-tumor activity. EXPERT OPINION There are a number of natural products or their derivatives that show excellent protection against cisplatin ototoxicity in preclinical studies. There is a need to insure uniform standards for purity of drugs derived from natural sources and to ensure adequate pharmacokinetics and safety of these products. Natural products that protect against cisplatin ototoxicity and augment cisplatin's anti-tumor effects in multiple studies of tumor-bearing animals are most promising for advancement to clinical trials. The most promising natural products include honokiol, sulforaphane, and thymoquinone.
Collapse
Affiliation(s)
- Leonard P Rybak
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Ian Alberts
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Shree Patel
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Raheem F H Al Aameri
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
6
|
Ezzeldeen Y, Swidan S, ElMeshad A, Sebak A. Green Synthesized Honokiol Transfersomes Relieve the Immunosuppressive and Stem-Like Cell Characteristics of the Aggressive B16F10 Melanoma. Int J Nanomedicine 2021; 16:5693-5712. [PMID: 34465990 PMCID: PMC8402984 DOI: 10.2147/ijn.s314472] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/17/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Honokiol (HK) is a natural bioactive compound with proven antineoplastic properties against melanoma. However, it shows very low bioavailability when administered orally. Alternatively, topical administration may offer a promising route. The objective of the current study was to fabricate HK transfersomes (HKTs) for topical treatment of melanoma. As an ultradeformable carrier system, transfersomes can overcome the physiological barriers to topical treatment of melanoma: the stratum corneum and the anomalous tumor microenvironment. Moreover, the immunomodulatory and stemness-regulation roles of HKTs were the main interest of this study. METHODS TFs were prepared using the modified scalable heating method. A three-factor, three-level Box-Behnken design was utilized for the optimization of the process and formulation variables. Intracellular uptake and cytotoxicity of HKTs were evaluated in nonactivated and stromal cell-activated B16F10 melanoma cells to investigate the influence of the complex tumor microenvironment on the efficacy of HK. Finally, ELISA and Western blot were performed to evaluate the expression levels of TGF-β and clusters of differentiation (CD47 and CD133, respectively). RESULTS The optimized formula exhibited a mean size of 190 nm, highly negative surface charge, high entrapment efficiency, and sustained release profile. HKTs showed potential to alleviate the immunosuppressive characteristics of B16F10 melanoma in vitro via downregulation of TGF-β signaling. In addition, HKTs reduced expression of the "do not eat me" signal - CD47. Moreover, HKTs possessed additional interesting potential to reduce the expression of the stem-like cell marker CD133. These outcomes were boosted upon combination with metformin, an antihyperglycemic drug recently reported to possess different functions in cancer, while combination with collagenase, an extracellular matrix-depleting enzyme, produced detrimental effects. CONCLUSION HKTs represent a promising scalable formulation for treatment of the aggressive B16F10 melanoma, which is jam-packed with immunosuppressive and stem-like cell markers.
Collapse
Affiliation(s)
- Yasmeen Ezzeldeen
- Department of Pharmaceutics, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo, 11837, Egypt
| | - Shady Swidan
- Department of Pharmaceutics, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo, 11837, Egypt
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt
| | - Aliaa ElMeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Bio Nano, Faculty of Nanotechnology for Postgraduate Studies, Cairo University, El-Sheikh Zayed, Giza, 12588, Egypt
| | - Aya Sebak
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo, Egypt
| |
Collapse
|
7
|
Counteracting Cisplatin-Induced Testicular Damages by Natural Polyphenol Constituent Honokiol. Antioxidants (Basel) 2020; 9:antiox9080723. [PMID: 32784851 PMCID: PMC7464045 DOI: 10.3390/antiox9080723] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
Cisplatin, despite its anti-cancer ability, exhibits severe testicular toxicities when applied systemically. Due to its wide application in cancer treatment, reduction of its damages to normal tissue is an imminent clinical need. Here we evaluated the effects of honokiol, a natural lipophilic polyphenol compound, on cisplatin-induced testicular injury. We showed in-vitro and in-vivo that nanosome-encapsulated honokiol attenuated cisplatin-induced DNA oxidative stress by suppressing intracellular reactive oxygen species production and elevating gene expressions of mitochondrial antioxidation enzymes. Nanosome honokiol also mitigated endoplasmic reticulum stress through down regulation of Bip-ATF4-CHOP signaling pathway. Additionally, this natural polyphenol compound diminished cisplatin-induced DNA breaks and cellular apoptosis. The reduced type I collagen accumulation in the testis likely attributed from inhibition of TGFβ1, αSMA and ER protein TXNDC5 protein expression. The combinatorial beneficial effects better preserve spermatogenic layers and facilitate repopulation of sperm cells. Our study renders opportunity for re-introducing cisplatin to systemic anti-cancer therapy with reduced testicular toxicity and restored fertility.
Collapse
|
8
|
Mukherjee A, Madamsetty VS, Paul MK, Mukherjee S. Recent Advancements of Nanomedicine towards Antiangiogenic Therapy in Cancer. Int J Mol Sci 2020; 21:E455. [PMID: 31936832 PMCID: PMC7013812 DOI: 10.3390/ijms21020455] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis is a process of generation of de-novo blood vessels from already existing vasculature. It has a crucial role in different physiological process including wound healing, embryonic development, and tumor growth. The methods by which therapeutic drugs inhibit tumor angiogenesis are termed as anti-angiogenesis cancer therapy. Developments of angiogenic inhibiting drugs have various limitations causing a barrier for successful treatment of cancer, where angiogenesis plays an important role. In this context, investigators developed novel strategies using nanotechnological approaches that have demonstrated inherent antiangiogenic properties or used for the delivery of antiangiogenic agents in a targeted manner. In this present article, we decisively highlight the recent developments of various nanoparticles (NPs) including liposomes, lipid NPs, protein NPs, polymer NPs, inorganic NPs, viral and bio-inspired NPs for potential application in antiangiogenic cancer therapy. Additionally, the clinical perspectives, challenges of nanomedicine, and future perspectives are briefly analyzed.
Collapse
Affiliation(s)
- Anubhab Mukherjee
- Aavishkar Oral Strips Pvt Ltd., 109/3, IDA, Phase 2, Sector 2, Lane 6, Cherlapally, Hyderabad 500051, India;
| | - Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - Manash K. Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, The University of California, Los Angeles (UCLA), Factor Bldg. 10-240, 621 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| |
Collapse
|
9
|
Eliaz I, Weil E. Intravenous Honokiol in Drug-Resistant Cancer: Two Case Reports. Integr Cancer Ther 2020; 19:1534735420922615. [PMID: 32482152 PMCID: PMC7268168 DOI: 10.1177/1534735420922615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Context: Long-term patient survival in cancer is affected by drug resistance. Honokiol (HNK) is a small-molecule polyphenol isolated from the bark and seed cones of Magnolia officinalis. HNK has been shown to enhance the effects of chemotherapy and inhibit drug resistance in preclinical models. HNK was well tolerated in multiple animal models when administered orally, intravenously (IV), and via intraperitoneal route. However, there are limited human data on the use of HNK in general, and specifically via IV (HNK-IV) in cancer. Objective: We aim to assess the efficacy, safety, and tolerability of HNK-IV in patients with drug-resistant tumors. Methods: This is a case study of 2 cancer patients who utilized HNK-IV as part of their cancer treatment regimen. The initial infusion of HNK was 10 mg/kg body weight, and subsequent treatments were increased up to 50 mg/kg according to individual tolerance, over 2 weeks. Results: Positive clinical response was achieved in both patients, including improved symptoms and quality of life. No serious adverse side effects occurred, and there were no adverse effects on laboratory parameters (complete blood count, kidney, and liver function). Transient sedation and minor nausea were noted and resolved postinfusion. Conclusions: This is the first report of HNK-IV in human patients. Given the positive clinical results, safety, and tolerability, the use of HNK-IV warrants further investigation regarding optimum formulation, and its use as adjunctive therapy in cancer patients.
Collapse
Affiliation(s)
- Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA, USA
| | - Elaine Weil
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA, USA
| |
Collapse
|
10
|
Ong CP, Lee WL, Tang YQ, Yap WH. Honokiol: A Review of Its Anticancer Potential and Mechanisms. Cancers (Basel) 2019; 12:E48. [PMID: 31877856 PMCID: PMC7016989 DOI: 10.3390/cancers12010048] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer is characterised by uncontrolled cell division and abnormal cell growth, which is largely caused by a variety of gene mutations. There are continuous efforts being made to develop effective cancer treatments as resistance to current anticancer drugs has been on the rise. Natural products represent a promising source in the search for anticancer treatments as they possess unique chemical structures and combinations of compounds that may be effective against cancer with a minimal toxicity profile or few side effects compared to standard anticancer therapy. Extensive research on natural products has shown that bioactive natural compounds target multiple cellular processes and pathways involved in cancer progression. In this review, we discuss honokiol, a plant bioactive compound that originates mainly from the Magnolia species. Various studies have proven that honokiol exerts broad-range anticancer activity in vitro and in vivo by regulating numerous signalling pathways. These include induction of G0/G1 and G2/M cell cycle arrest (via the regulation of cyclin-dependent kinase (CDK) and cyclin proteins), epithelial-mesenchymal transition inhibition via the downregulation of mesenchymal markers and upregulation of epithelial markers. Additionally, honokiol possesses the capability to supress cell migration and invasion via the downregulation of several matrix-metalloproteinases (activation of 5' AMP-activated protein kinase (AMPK) and KISS1/KISS1R signalling), inhibiting cell migration, invasion, and metastasis, as well as inducing anti-angiogenesis activity (via the down-regulation of vascular endothelial growth factor (VEGFR) and vascular endothelial growth factor (VEGF)). Combining these studies provides significant insights for the potential of honokiol to be a promising candidate natural compound for chemoprevention and treatment.
Collapse
Affiliation(s)
| | | | - Yin Quan Tang
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University Lakeside Campus, No. 1, Jalan Taylor’s, Subang Jaya 47500, Malaysia; (C.P.O.); (W.L.L.)
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University Lakeside Campus, No. 1, Jalan Taylor’s, Subang Jaya 47500, Malaysia; (C.P.O.); (W.L.L.)
| |
Collapse
|
11
|
Zhu M, Li B, Ma H, Huang X, Wang H, Dai Y, Li Y, Li HM, Wu CZ. Synthesis and in vitro antitumor evaluation of honokiol derivatives. Bioorg Med Chem Lett 2019; 30:126849. [PMID: 31831382 DOI: 10.1016/j.bmcl.2019.126849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 11/29/2022]
Abstract
Honokiol is a natural bioactive neolignan and has been widely researched and structural modified as an anticancer agent. In this paper, 18 honokiol derivatives were synthesized and investigated for their antitumor activity. Among these, the promising compound 5a exhibited much higher anti-proliferative activity with IC50 value of 10.41 μM. Transwell assays showed that 5a could significantly inhibit the invasion and migration of I-10 cells at 2.5 μM, which was further confirmed by the western blotting experiments with down-regulation of the HIF-1α and its associated downstream proteins MMP-2 and MMP-9. Overall, these results provided useful suggestion for further structural optimization of honokiol derivatives.
Collapse
Affiliation(s)
- Meilin Zhu
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China
| | - Bohan Li
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China
| | - Hui Ma
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China
| | - Xuenan Huang
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China
| | - Haotian Wang
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China
| | - Yiqun Dai
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China
| | - Yu Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Hong-Mei Li
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China.
| | - Cheng-Zhu Wu
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, Anhui, China.
| |
Collapse
|
12
|
Eliaz I, Raz A. Pleiotropic Effects of Modified Citrus Pectin. Nutrients 2019; 11:nu11112619. [PMID: 31683865 PMCID: PMC6893732 DOI: 10.3390/nu11112619] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/24/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Modified citrus pectin (MCP) has a low-molecular-weight degree of esterification to allow absorption from the small intestinal epithelium into the circulation. MCP produces pleiotropic effects, including but not limited to its antagonism of galectin-3, which have shown benefit in preclinical and clinical models. Regarding cancer, MCP modulates several rate-limiting steps of the metastatic cascade. MCP can also affect cancer cell resistance to chemotherapy. Regarding fibrotic diseases, MCP modulates many of the steps involved in the pathogenesis of aortic stenosis. MCP also reduces fibrosis to the kidney, liver, and adipose tissue. Other benefits of MCP include detoxification and improved immune function. This review summarizes the pleiotropic effects of MCP.
Collapse
Affiliation(s)
- Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, 398 Tesconi Ct, Santa Rosa, CA 95401, USA.
| | - Avraham Raz
- Departments of Oncology and Pathology, School of Medicine, Wayne State University and Barbara Ann Karmanos Cancer Institute, 4100 John R St, Detroit, MI 48201, USA.
| |
Collapse
|
13
|
Liu HT, Wang TE, Hsu YT, Chou CC, Huang KH, Hsu CC, Liang HJ, Chang HW, Lee TH, Tsai PS. Nanoparticulated Honokiol Mitigates Cisplatin-Induced Chronic Kidney Injury by Maintaining Mitochondria Antioxidant Capacity and Reducing Caspase 3-Associated Cellular Apoptosis. Antioxidants (Basel) 2019; 8:antiox8100466. [PMID: 31600935 PMCID: PMC6826708 DOI: 10.3390/antiox8100466] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is a potent anti-cancer drug, however, its accompanied organ-toxicity hampers its clinical applications. Cisplatin-associated kidney injury is known to result from its accumulation in the renal tubule with excessive generation of reactive oxygen species. In this study, we encapsulated honokiol, a natural lipophilic polyphenol constituent extracted from Magnolia officinalis into nano-sized liposomes (nanosome honokiol) and examined the in vivo countering effects on cisplatin-induced renal injury. We observed that 5 mg/kg body weight. nanosome honokiol was the lowest effective dosage to efficiently restore renal functions of cisplatin-treated animals. The improvement is likely due the maintenance of cellular localization of cytochrome c and thus preserves mitochondria integrity and their redox activity, which as a consequence, reduced cellular oxidative stress and caspase 3-associated apoptosis. These improvements at the cellular level are later reflected on the observed reduction of kidney inflammation and fibrosis. In agreement with our earlier in vitro study showing protective effects of honokiol on kidney cell lines, we demonstrated further in the current study, that nanosuspension-formulated honokiol provides protective effects against cisplatin-induced chronic kidney damages in vivo. Our findings not only benefit cisplatin-receiving patients with reduced renal side effects, but also provide potential alternative and synergic solutions to improve clinical safety and efficacy of cisplatin treatment on cancer patients.
Collapse
Affiliation(s)
- Hung-Ting Liu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Tse-En Wang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Yu-Ting Hsu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Chi-Chung Chou
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, 402 Taichung, Taiwan.
| | - Kai-Hung Huang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Hong-Jen Liang
- Department of Food Science, Yuanpei University, 30015 Hsinchu, Taiwan.
| | - Hui-Wen Chang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Tzong-Huei Lee
- Institute of Fisheries Science, National Taiwan University, Taipei 10617, Taiwan.
| | - Pei-Shiue Tsai
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
14
|
Lin D, Yan Z, Chen A, Ye J, Hu A, Liu J, Peng J, Wu X. Anti-proliferative activity and structure-activity relationship of honokiol derivatives. Bioorg Med Chem 2019; 27:3729-3734. [PMID: 31278004 DOI: 10.1016/j.bmc.2019.06.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 12/27/2022]
Abstract
As a known natural product with anti-tumor activity, honokiol has been widely researched and structural modified. Lots of honokiol derivatives have been found to possess good anti-proliferative activity and showed great potential in cancer therapy, but the SAR (structure-activity relationship) was still confused. Here in, the SAR were comprehensively researched by summary of reported derivatives and synthesis of novel derivatives. Amongst novel derivatives, the promising compounds A6 and A10 exhibited potent and selective anti-proliferative activities against K562 cell line with the IC50 values of 5.04 and 7.08 μM respectively. The SAR was discussed around honokiol and 79 derivatives by the means of CoMFA and theoretical calculation, which provided useful suggestion for further structural optimization of honokiol derivatives.
Collapse
Affiliation(s)
- Ding Lin
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China; Department of Traditional Chinese Medicine, Zhejiang A & F University, Hangzhou 311300, China
| | - Zhongzhong Yan
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Aiyu Chen
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jiao Ye
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Aixi Hu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Juan Liu
- College of Pharmacy and Biological Science, University of South China, Hengyang 421000, China
| | - Junmei Peng
- College of Pharmacy and Biological Science, University of South China, Hengyang 421000, China
| | - Xiaoyun Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
15
|
Banik K, Ranaware AM, Deshpande V, Nalawade SP, Padmavathi G, Bordoloi D, Sailo BL, Shanmugam MK, Fan L, Arfuso F, Sethi G, Kunnumakkara AB. Honokiol for cancer therapeutics: A traditional medicine that can modulate multiple oncogenic targets. Pharmacol Res 2019; 144:192-209. [DOI: 10.1016/j.phrs.2019.04.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/18/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
|
16
|
Lee JS, Sul JY, Park JB, Lee MS, Cha EY, Ko YB. Honokiol induces apoptosis and suppresses migration and invasion of ovarian carcinoma cells via AMPK/mTOR signaling pathway. Int J Mol Med 2019; 43:1969-1978. [PMID: 30864681 PMCID: PMC6443331 DOI: 10.3892/ijmm.2019.4122] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 02/18/2019] [Indexed: 01/08/2023] Open
Abstract
Honokiol, a natural biphenolic compound, exerts anticancer effects through a variety of mechanisms on multiple types of cancer with relatively low toxicity. Adenosine 5'‑phosphate‑activated protein kinase (AMPK), an essential regulator of cellular homeostasis, may control cancer progression. The present study aimed to investigate whether the anticancer activities of honokiol in ovarian cancer cells were mediated through the activation of AMPK. Honokiol decreased cell viability of 2 ovarian cancer cell lines, with an half‑maximal inhibitory concentration value of 48.71±11.31 µM for SKOV3 cells and 46.42±5.37 µM for Caov‑3 cells. Honokiol induced apoptosis via activation of caspase‑3, caspase‑7 and caspase‑9, and cleavage of poly‑(adenosine 5'‑diphosphate‑ribose) polymerase. Apoptosis induced by honokiol was weakened by compound C, an AMPK inhibitor, suggesting that honokiol‑induced apoptosis was dependent on the AMPK/mechanistic target of rapamycin signaling pathway. Additionally, honokiol inhibited the migration and invasion of ovarian cancer cells. The combined treatment of honokiol with compound C reversed the activities of honokiol in wound healing and Matrigel invasion assays. These results indicated that honokiol may have therapeutic potential in ovarian cancer by targeting AMPK activation.
Collapse
Affiliation(s)
- Jin Sun Lee
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Ji Young Sul
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jun Beom Park
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Myung Sun Lee
- Surgical Oncology Research Laboratory, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Eun Young Cha
- Surgical Oncology Research Laboratory, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Young Bok Ko
- Research Institute for Medicinal Sciences, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
17
|
Rauf A, Patel S, Imran M, Maalik A, Arshad MU, Saeed F, Mabkhot YN, Al-Showiman SS, Ahmad N, Elsharkawy E. Honokiol: An anticancer lignan. Biomed Pharmacother 2018; 107:555-562. [DOI: 10.1016/j.biopha.2018.08.054] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/20/2018] [Accepted: 08/10/2018] [Indexed: 01/22/2023] Open
|
18
|
AbdElhamid AS, Zayed DG, Helmy MW, Ebrahim SM, Bahey-El-Din M, Zein-El-Dein EA, El-Gizawy SA, Elzoghby AO. Lactoferrin-tagged quantum dots-based theranostic nanocapsules for combined COX-2 inhibitor/herbal therapy of breast cancer. Nanomedicine (Lond) 2018; 13:2637-2656. [DOI: 10.2217/nnm-2018-0196] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Herein, tumor-targeted quantum dots (QDs)-based theranostic nanocapsules (NCs) coloaded with celecoxib and honokiol were developed. Materials & methodology: The anionic CD44-targeting chondroitin sulfate and cationic low density lipoprotein (LDL)-targeting lactoferrin (LF) were sequentially assembled onto the surface of the positively charged oily core. As an imaging probe, highly fluorescent mercaptopropionic acid-capped cadmium telluride QDs were coupled to LF. Results: In vitro, fluorescence of QDs was quenched (OFF state) due to combined electron/energy transfer-mediated processes involving LF. After intracellular uptake of NCs, fluorescence was restored (ON state), thus enabled tracing their internalization. The NCs demonstrated enhanced cytotoxicity against breast cancer cells as well as superior in vivo antitumor efficacy. Conclusion: We propose these multifunctional nanotheranostics for imaging and targeted therapy of breast cancer.
Collapse
Affiliation(s)
- Ahmed S AbdElhamid
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Dina G Zayed
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Maged W Helmy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Damanhour University, El Bahira, Egypt
| | - Shaker M Ebrahim
- Department of Materials Science, Institute of Graduate Studies & Research, Alexandria University, Alexandria, Egypt
| | - Mohammed Bahey-El-Din
- Department of Microbiology & Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Esmat A Zein-El-Dein
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Sanaa A El-Gizawy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA
| |
Collapse
|
19
|
Achkar IW, Abdulrahman N, Al-Sulaiti H, Joseph JM, Uddin S, Mraiche F. Cisplatin based therapy: the role of the mitogen activated protein kinase signaling pathway. J Transl Med 2018; 16:96. [PMID: 29642900 PMCID: PMC5896132 DOI: 10.1186/s12967-018-1471-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/19/2022] Open
Abstract
Cisplatin is a widely used chemotherapeutic agent for treatment of various cancers. However, treatment with cisplatin is associated with drug resistance and several adverse side effects such as nephrotoxicity, reduced immunity towards infections and hearing loss. A Combination of cisplatin with other drugs is an approach to overcome drug resistance and reduce toxicity. The combination therapy also results in increased sensitivity of cisplatin towards cancer cells. The mitogen activated protein kinase (MAPK) pathway in the cell, consisting of extracellular signal regulated kinase, c-Jun N-terminal kinase, p38 kinases, and downstream mediator p90 ribosomal s6 kinase (RSK); is responsible for the regulation of various cellular events including cell survival, cell proliferation, cell cycle progression, cell migration and protein translation. This review article demonstrates the role of MAPK pathway in cisplatin based therapy, illustrates different combination therapy involving cisplatin and also shows the importance of targeting MAPK family, particularly RSK, to achieve increased anticancer effect and overcome drug resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Iman W Achkar
- Translational Research Institute, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | | | - Hend Al-Sulaiti
- College of Pharmacy, Qatar University, P.O. Box 2713, Doha, Qatar
| | | | - Shahab Uddin
- Translational Research Institute, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Fatima Mraiche
- College of Pharmacy, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
20
|
Huang KJ, Kuo CH, Chen SH, Lin CY, Lee YR. Honokiol inhibits in vitro and in vivo growth of oral squamous cell carcinoma through induction of apoptosis, cell cycle arrest and autophagy. J Cell Mol Med 2018; 22:1894-1908. [PMID: 29363886 PMCID: PMC5824386 DOI: 10.1111/jcmm.13474] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/30/2017] [Indexed: 01/22/2023] Open
Abstract
Honokiol, an active natural product derived from Magnolia officinalis, exerted anticancer effects through a variety of mechanisms on multiple types of cancers. In this study, the molecular mechanisms of honokiol in suppressing the human oral squamous cell carcinoma (OSCC) cells were evaluated. Treatment of two OSCC cell lines with honokiol resulted in reducing the cell proliferation and arresting the cell cycle at G1 stage which was correlated with the down‐regulation of Cdk2 and Cdk4 and the up‐regulation of cell cycle suppressors, p21 and p27. In addition, the caspase‐dependent programmed cell death was substantially detected, and the autophagy was induced as the autophagosome formation and autophagic flux proceeded. Modulation of autophagy by autophagic inducer, rapamycin or inhibitors, 3‐MA or bafilomycin, potentiated the honokiol‐mediated anti‐OSCC effects where honokiol exerted multiple actions in suppression of MAPK pathway and regulation of Akt/mTOR or AMPK pathways. As compared to clinical therapeutic agent, 5‐FU, honokiol exhibited more potent activity against OSCC cells and synergistically enhanced the cytotoxic effect of 5‐FU. Furthermore, orally administrated honokiol exerted effective antitumour activity in vivo in OSCC‐xenografted mice. Thus, this study revealed that honokiol could be a promising candidate in preventing human OSCCs.
Collapse
Affiliation(s)
- Kao-Jean Huang
- Development Center for Biotechnology, Institute of Biologics, New Taipei City, Taiwan
| | - Chin-Ho Kuo
- Division of Hematology-Oncology and Blood Bank, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Shu-Hsin Chen
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Ching-Yen Lin
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Ying-Ray Lee
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan.,Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan
| |
Collapse
|
21
|
Antineoplastic Effects of Honokiol on Melanoma. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5496398. [PMID: 28194418 PMCID: PMC5282456 DOI: 10.1155/2017/5496398] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022]
Abstract
Honokiol, a plant lignan has been shown to have antineoplastic effects against nonmelanoma skin cancer developments in mice. In this study, antineoplastic effects of honokiol were investigated in malignant melanoma models. In vitro effects of honokiol treatment on SKMEL-2 and UACC-62 melanoma cells were evaluated by measuring the cell viability, proliferation, apoptosis, cell cycle analysis, and expressions of various proteins associated with cell cycle progression and apoptosis. For the in vivo study, male nude mice inoculated with SKMEL-2 or UACC-62 cells received injections of sesame oil or honokiol for two to seven weeks. In vitro honokiol treatment caused significant decrease in cell viability, proliferation, cell cycle arrest, increased apoptosis, and modulation of apoptotic and cell cycle regulatory proteins. Honokiol caused an accumulation of cells in the G2/M phase of the cell cycle in SKMEL-2 and G0/G1 phase in UACC-62 cells. An elevated level of caspases and PARP were observed in both cell lines treated with honokiol. A decrease in the expression of various cell cycle regulatory proteins was also observed in honokiol treated cells. Honokiol caused a significant reduction of tumor growth in SKMEL-2 and UACC-62 melanoma xenografts. These findings suggest that honokiol is a good candidate for further studies as a possible treatment for malignant melanoma.
Collapse
|
22
|
Yang J, Pei H, Luo H, Fu A, Yang H, Hu J, Zhao C, Chai L, Chen X, Shao X, Wang C, Wu W, Wan L, Ye H, Qiu Q, Peng A, Wei Y, Yang L, Chen L. Non-toxic dose of liposomal honokiol suppresses metastasis of hepatocellular carcinoma through destabilizing EGFR and inhibiting the downstream pathways. Oncotarget 2016; 8:915-932. [PMID: 27906672 PMCID: PMC5352206 DOI: 10.18632/oncotarget.13687] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/05/2016] [Indexed: 02/05/2023] Open
Abstract
At present, there is no specific anti-metastasis drug in HCC treatment. Drugs used for primary HCC tumors and tumor metastasis are very similar, among which cytotoxic drugs are prevalent, such as cisplatin, doxorubicin and 5-FU. The EGFR pathway plays an important role in promoting hepatocellular carcinoma (HCC) metastasis. Hence, development of non-toxic anti-metastasis drugs, such as EGFR or downstream pathways inhibitors, is of great importance. In our present study, we found non-toxic dose of liposomal honokiol (LH) could inhibit the HCC metastasis by destabilizing EGFR and inhibiting the downstream pathways. Non-toxic dose of LH significantly inhibited the motility, migration and lamellipodia formation of HepG2 cells in vitro and decreased extravasation of HepG2 cells in a novel metastasis model of transgenic zebrafish. In two lung metastasis models (HepG2 and B16F10) and a spontaneous metastasis model of HepG2 cells, LH remarkably inhibited pulmonary metastasis and regional lymph nodes metastasis without obvious toxicity. Further study showed that destabilizing EGFR and inhibiting the downstream pathways were the main mechanisms of non-toxic dose of LH on metastasis inhibition. Our results provide the preclinical rationale and the underlying mechanisms of LH to suppress HCC metastasis, implicating LH as a potential therapeutic agent to block HCC metastasis without severe side effects.
Collapse
Affiliation(s)
- Jianhong Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Heying Pei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Hong Luo
- 2 Department of Ultrasonic Medicine, West China Second Hospital, Sichuan University, Chengdu, China
| | - Afu Fu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Hansuo Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Jia Hu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Chengjian Zhao
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - LuLu Chai
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Xiang Chen
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Ximing Shao
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Chunyu Wang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Wenshuang Wu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Li Wan
- 3 School of Pharmacy, Chengdu University of TCM, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Haoyu Ye
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Qiang Qiu
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Aihua Peng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Yuquan Wei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Li Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Lijuan Chen
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| |
Collapse
|
23
|
Ji N, Jiang L, Deng P, Xu H, Chen F, Liu J, Li J, Liao G, Zeng X, Lin Y, Feng M, Li L, Chen Q. Synergistic effect of honokiol and 5-fluorouracil on apoptosis of oral squamous cell carcinoma cells. J Oral Pathol Med 2016; 46:201-207. [PMID: 27465776 DOI: 10.1111/jop.12481] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Ning Ji
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Peng Deng
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Hao Xu
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Fangman Chen
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Jinli Liu
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Jing Li
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Ga Liao
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Yuchun Lin
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Mingye Feng
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan China
| |
Collapse
|
24
|
Hahm ER, Singh KB, Singh SV. c-Myc is a novel target of cell cycle arrest by honokiol in prostate cancer cells. Cell Cycle 2016; 15:2309-20. [PMID: 27341160 DOI: 10.1080/15384101.2016.1201253] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Honokiol (HNK), a highly promising phytochemical derived from Magnolia officinalis plant, exhibits in vitro and in vivo anticancer activity against prostate cancer but the underlying mechanism is not fully clear. This study was undertaken to delineate the role of c-Myc in anticancer effects of HNK. Exposure of prostate cancer cells to plasma achievable doses of HNK resulted in a marked decrease in levels of total and/or phosphorylated c-Myc protein as well as its mRNA expression. We also observed suppression of c-Myc protein in PC-3 xenografts upon oral HNK administration. Stable overexpression of c-Myc in PC-3 and 22Rv1 cells conferred significant protection against HNK-mediated growth inhibition and G0-G1 phase cell cycle arrest. HNK treatment decreased expression of c-Myc downstream targets including Cyclin D1 and Enhancer of Zeste Homolog 2 (EZH2), and these effects were partially restored upon c-Myc overexpression. In addition, PC-3 and DU145 cells with stable knockdown of EZH2 were relatively more sensitive to growth inhibition by HNK compared with control cells. Finally, androgen receptor overexpression abrogated HNK-mediated downregulation of c-Myc and its targets particularly EZH2. The present study indicates that c-Myc, which is often overexpressed in early and late stages of human prostate cancer, is a novel target of prostate cancer growth inhibition by HNK.
Collapse
Affiliation(s)
- Eun-Ryeong Hahm
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA.,b University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA
| | - Krishna Beer Singh
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA.,b University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA
| | - Shivendra V Singh
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA.,b University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA
| |
Collapse
|
25
|
Zuo GY, Zhang XJ, Han J, Li YQ, Wang GC. In vitro synergism of magnolol and honokiol in combination with antibacterial agents against clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA). BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:425. [PMID: 26627468 PMCID: PMC4666064 DOI: 10.1186/s12906-015-0938-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/17/2015] [Indexed: 02/03/2023]
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is a problematic pathogen posing a serious therapeutic challenge in the clinic. It is often multidrug-resistant (MDR) to conventional classes of antibacterial agents and there is an urgent need to develop new agents or strategies for treatment. Magnolol (ML) and honokiol (HL) are two naturally occurring diallylbiphenols which have been reported to show inhibition of MRSA. In this study their synergistic effects with antibacterial agents were further evaluated via checkerboard and time-kill assays. Methods The susceptibility spectrum of clinical MRSA strains was tested by the disk diffusion method. The minimal inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) of ML and HL were assayed by broth microdilution. The synergy was evaluated through checkerboard microdilution and time-killing experiments. Results ML and HL showed similar activity against both MSSA and MRSA with MIC/MBC at 16 ~ 64 mg/L, with potency similar to amikacin (AMK) and gentamicin (GEN). When they were used in combination with conventional antibacterial agents, they showed bacteriostatic synergy with FICIs between 0.25 ~ 0.5, leading to the combined MICs decreasing to as low as 1 ~ 2 and 1 ~ 16 mg/L for ML (HL) and the agents, respectively. MIC50 of the combinations decreased from 16 mg/L to 1 ~ 4 mg/L for ML (HL) and 8 ~ 128 mg/L to 2 ~ 64 mg/L for the antibacterial agents, which exhibited a broad spectrum of synergistic action with aminoglycosides (AMK, etilmicin (ETM) and GEN), floroquinolones (levofloxacin (LEV), ciprofloxacin and norfloxacin), fosfomycin (FOS) and piperacillin. The times of dilution (TOD, the extent of decreasing in MIC value) were determined up to 16 for the combined MIC. A more significant synergy after combining was determined as ML (HL) with AMK, ETM, GEN and FOS. ML (HL) combined with antibacterial agents did not show antagonistic effects on any of the ten MRSA strains. Reversal effects of MRSA resistance to AMK and GEN by ML and HL were also observed, respectively. All the combinations also showed better dynamic bactericidal activity against MRSA than any of single ML (HL) or the agents at 24 h incubation. The more significant synergy of combinations were determined as HL (ML) + ETM, HL + LEV and HL + AMK (GEN or FOS), with △LC24 of 2.02 ~ 2.25. Conclusion ML and HL showed synergistic potentiation of antibacterial agents against clinical isolates of MRSA and warrant further pharmacological investigation.
Collapse
|
26
|
Hahm ER, Karlsson AI, Bonner MY, Arbiser JL, Singh SV. Honokiol inhibits androgen receptor activity in prostate cancer cells. Prostate 2014; 74:408-20. [PMID: 24338950 PMCID: PMC3946953 DOI: 10.1002/pros.22762] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 11/20/2013] [Indexed: 01/19/2023]
Abstract
BACKGROUND We have shown previously that honokiol (HNK), a bioactive component of the medicinal plant Magnolia officinalis, inhibits growth of human prostate cancer cells in vitro and in vivo. However, the effect of HNK on androgen receptor (AR) signaling has not been studied. METHODS LNCaP, C4-2, and TRAMP-C1 cells were used for various assays. Trypan blue dye exclusion assay or clonogenic assay was performed for determination of cell viability. The effects of HNK and/or its analogs on protein levels of AR and its target gene product prostate specific antigen (PSA) were determined by western blotting. RNA interference of p53 was achieved by transient transfection. Reverse transcription-polymerase chain reaction was performed for mRNA expression of AR. Nuclear level of AR was visualized by microscopy. Apoptosis was quantified by DNA fragmentation assay or flow cytometry after Annexin V-propidium iodide staining. RESULTS HNK and its dichloroacetate analog (HDCA) were relatively more effective in suppressing cell viability and AR protein level than honokiol epoxide or biseugenol. Nuclear translocation of AR stimulated by a synthetic androgen (R1881) was markedly suppressed in the presence of HNK. Downregulation of AR protein resulting from HNK exposure was attributable to transcriptional repression as well as proteasomal degradation. HNK-mediated suppression of AR protein was maintained in LNCaP cells after knockdown of p53 protein. HNK-induced apoptosis was not affected by R1881 treatment. CONCLUSIONS The present study demonstrates, for the first time, that HNK inhibits activity of AR in prostate cancer cells regardless of the p53 status.
Collapse
Affiliation(s)
- Eun-Ryeong Hahm
- Department of Pharmacology & Chemical Biology, and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. Isabella Karlsson
- Department of Dermatology and Atlanta Veterans Administration Medical Center, Emory University School of Medicine, Atlanta, GA
| | - Michael Y. Bonner
- Department of Dermatology and Atlanta Veterans Administration Medical Center, Emory University School of Medicine, Atlanta, GA
| | - Jack L. Arbiser
- Department of Dermatology and Atlanta Veterans Administration Medical Center, Emory University School of Medicine, Atlanta, GA
- Winship Cancer Institute, Emory University, Atlanta, GA
| | - Shivendra V. Singh
- Department of Pharmacology & Chemical Biology, and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Correspondence to: Shivendra V. Singh, 2.32A Hillman Cancer Center Research Pavilion, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213. Phone: 412-623-3263; Fax: 412-623-7828;
| |
Collapse
|
27
|
Honokiol analogs: a novel class of anticancer agents targeting cell signaling pathways and other bioactivities. Future Med Chem 2013; 5:809-29. [PMID: 23651094 DOI: 10.4155/fmc.13.32] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Honokiol (3,5-di-(2-propenyl)-1,1-biphenyl-2,2-diol) is a natural bioactive neolignan isolated from the genus Magnolia. In recent studies, honokiol has been observed to have anti-angiogenic, anticancer, anti-inflammatory, neuroprotective and GABA-modulating properties in vitro and in preclinical models. Honokiol and its analogs target multiple signaling pathways including NF-κB, STAT3, EGFR, mTOR and caspase-mediated common pathway, which regulate cancer initiation and progression. Honokiol and its targets of action may be helpful in the development of effective analogs and targeted cancer therapy. In this review, recent data describing the molecular targets of honokiol and its analogs with anticancer and some other bioactivities are discussed.
Collapse
|
28
|
He Z, Subramaniam D, Zhang Z, Zhang Y, Anant S. Honokiol as a Radiosensitizing Agent for Colorectal cancers. CURRENT COLORECTAL CANCER REPORTS 2013; 9. [PMID: 24307888 DOI: 10.1007/s11888-013-0191-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Radioresistance is a frustrating obstacle for patients with colorectal cancers (CRCs) undergoing radiotherapy. There is an urgent need to find an effective agent to increase the sensitivity of CRCs to radiation. Honokiol, an active compound purified from Magnolia, was found to radiosensitize colorectal cancer cells both in vitro and in vivo. However, the mechanisms control important signaling that enhances radiosensitivity is currently unknown. In this study, we have reviewed important signaling pathways that are closely related to radiosensitization, such as cell cycle arrest, tumor angiogenesis, JAK/STAT3 signaling pathway and Mismatch repair. Studies show that honokiol can interfere with these pathways at different levels. With overall analysis, it may bring light on finding the possible mechanism by which honokiol acts as a radiosensitizing agent for CRCs.
Collapse
Affiliation(s)
- Zhiyun He
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China ; Department of General Surgery, Second Hospital of Lanzhou, University of Lanzhou, Gansu 730030, China ; Department of Molecular and Integrative Physiology, Kansas City, Kansas, USA
| | | | | | | | | |
Collapse
|
29
|
Identification of metabolites of honokiol in rat urine using 13C stable isotope labeling and liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry. J Chromatogr A 2013; 1295:48-56. [DOI: 10.1016/j.chroma.2013.03.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 03/09/2013] [Accepted: 03/12/2013] [Indexed: 12/27/2022]
|
30
|
Pan XY, Li X, Che YC, Li HY, Li X, Zhang Y, Yang X. Overexpression of claudin-4 may be involved in endometrial tumorigenesis. Oncol Lett 2013; 5:1422-1426. [PMID: 23599806 PMCID: PMC3628940 DOI: 10.3892/ol.2013.1198] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 02/07/2013] [Indexed: 01/11/2023] Open
Abstract
To clarify the role of claudin-4 in endometrial tumorigenesis and to explore whether claudin-4 could be a potentially useful agent in the treatment of endometrial carcinoma, the expression of claudin-4 in endometrial carcinoma was investigated. The relationship between therapy with anti-neoplastic agents and the expression of claudin-4 was also analyzed using an endometrial carcinoma xenograft model. The expression of claudin-4 in endometrial endometrioid adenocarcinoma (EEC) and normal human endometrial tissue was determined using immunohistochemistry and real-time PCR. Ninety female BALB/c nu/nu mice were transplanted with Ishikawa endometrial cancer cells. The mice were divided into three groups with different intraperitoneal treatments: cisplatin, paclitaxel or saline solution. After the observation period tumors were extracted and stained with monoclonal antibody against claudin-4. The mRNA expression of claudin-4 was also detected using real-time PCR. Expression of claudin-4 was significantly increased at both protein and mRNA levels in the EEC group compared with the group of normal cyclic endometrium. In the study of Ishikawa xenografts, no significant changes in tumor volume and claudin-4 expression were shown in the paclitaxel group compared with the control group. A significant reduction of tumor growth and a significant decrease in claudin-4 expression were observed in the cisplatin group. These results demonstrate that claudin-4 is strongly expressed in EEC. Claudin-4 is a useful biomarker in the treatment of patients with endometrial carcinoma.
Collapse
Affiliation(s)
- Xiao-Yu Pan
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing 100029
| | | | | | | | | | | | | |
Collapse
|
31
|
Zahedi P, Yoganathan R, Piquette-Miller M, Allen C. Recent advances in drug delivery strategies for treatment of ovarian cancer. Expert Opin Drug Deliv 2012; 9:567-83. [DOI: 10.1517/17425247.2012.665366] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
32
|
Kim JH, Jeong SJ, Kim B, Yun SM, Choi DY, Kim SH. Melatonin synergistically enhances cisplatin-induced apoptosis via the dephosphorylation of ERK/p90 ribosomal S6 kinase/heat shock protein 27 in SK-OV-3 cells. J Pineal Res 2012; 52:244-52. [PMID: 22050627 DOI: 10.1111/j.1600-079x.2011.00935.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
To evaluate melatonin's ability to enhance ovarian cancer cells to cisplatin treatment for ovarian cancer, this study was performed. Melatonin by itself had no significant cytotoxicity against SK-OV-3 cells, while cisplatin suppressed the cell viability in a dose-dependent manner. Combined treatment with cisplatin and melatonin synergistically inhibited the viability of SK-OV-3 cells with the synergism between two drugs (1 > combination index). In contrast, melatonin revealed the protective effect against cisplatin-induced cytotoxicity in OSEN normal ovarian epithelial cells. Cotreatment with cisplatin and melatonin increased the sub-G1 DNA contents and TdT-mediated dUTP nick end-labeling (TUNEL)-positive cells compared with cisplatin control in SK-OV-3 cells, suggesting that melatonin augments cisplatin-induced apoptosis. Consistently, combined treatment of cisplatin and melatonin increased the cleavage of caspase-3 and poly-(ADP-ribose) polymerase (PARP). Importantly, melatonin synergistically inhibited the phosphorylation of extracellular signal-regulated kinase (ERK) along with dephosphorylation of 90-kDa ribosomal S6 kinase (p90RSK) and heat shock protein 27 (HSP27) induced by cisplatin. Furthermore, melatonin remarkably blocked the expression and colocalization of p90RSK and HSP27 by combination treatment with cisplatin. Taken together, our findings demonstrate that melatonin enhances cisplatin-induced apoptosis via the inactivation of ERK/p90RSK/HSP27 cascade in SK-OV-3 cells as a potent synergist to cisplatin treatment.
Collapse
Affiliation(s)
- Ji-Hyun Kim
- College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
33
|
Xie Y, Long Q, Wu Q, Shi S, Dai M, Liu Y, Liu L, Gong C, Qian Z, Wei Y, Zhao X. Improving therapeutic effect in ovarian peritoneal carcinomatosis with honokiol nanoparticles in a thermosensitive hydrogel composite. RSC Adv 2012. [DOI: 10.1039/c2ra20612a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
34
|
Li CY, Chao LK, Wang SC, Chang HZ, Tsai ML, Fang SH, Liao PC, Ho CL, Chen ST, Cheng WC, Chiang CS, Kuo YH, Hua KF, Hsu IC. Honokiol inhibits LPS-induced maturation and inflammatory response of human monocyte-derived dendritic cells. J Cell Physiol 2011; 226:2338-49. [DOI: 10.1002/jcp.22576] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
35
|
Liang S, Fu A, Zhang Q, Tang M, Zhou J, Wei Y, Chen L. Honokiol inhibits HepG2 migration via down-regulation of IQGAP1 expression discovered by a quantitative pharmaceutical proteomic analysis. Proteomics 2010; 10:1474-83. [PMID: 20127691 DOI: 10.1002/pmic.200900649] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Honokiol (HNK), a natural small molecular product, inhibited proliferation of HepG2 cells and exhibited anti-tumor activity in nude mice. In this article, we applied a novel sensitive stable isotope labeling with amino acids in cell culture-based quantitative proteomic method and a model of nude mice to investigate the correlation between HNK and the hotspot migration molecule Ras GTPase-activating-like protein (IQGAP1). The quantitative proteomic analysis showed that IQGAP1 was 0.53-fold down-regulated under 10 microg/mL HNK exposure for 24 h on HepG2 cells. Migration ability of HepG2 cells under HNK treatment was correlated with its expression level of IQGAP1. In addition, the biochemical validation on HepG2 cells and the tumor xenograft model further demonstrated that HNK decreased the expression level of IQGAP1 and its upstream proteins Cdc42/Rac1. These data supported that HNK can modulate cell adhesion and cell migration by acting on Cdc42/Rac1 signaling via IQGAP1 interactions with its upstream Cdc42/Rac1 proteins, which is a new molecular mechanism of HNK to exert its anti-tumor activity.
Collapse
Affiliation(s)
- Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, West China Medical School, Sichuan University, Chengdu, PR China
| | | | | | | | | | | | | |
Collapse
|
36
|
Luo Y, Xu Y, Chen L, Hu J, Peng C, Xie D, Shi J, Huang W, Xu G, Peng M, Han J, Li R, Yang S, Wei Y. Semi-synthesis and anti-proliferative activity evaluation of novel analogues of Honokiol. Bioorg Med Chem Lett 2009; 19:4702-5. [PMID: 19589678 DOI: 10.1016/j.bmcl.2009.06.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 06/02/2009] [Accepted: 06/18/2009] [Indexed: 02/05/2023]
Abstract
A series of honokiol analogues were synthesized by modifying the 5- and/or 3'-position(s) of honokiol to assess their anti-tumor effects. Some compounds exerted more potent anti-proliferative activities than those of honokiol on K562 leukemia cells, A549 alveolar basal epithelial cells, SPC-A1 adenocarcinoma cells and A2780 human ovarian carcinoma cells in vitro. Compounds 2b, 3a, and 3c displayed most potent anti-proliferative activities against these tested cell strains and their anti-drug resistance effects were evaluated in vitro on cisplatin-resistant A2780 human ovarian carcinoma cells. The structure-activity relationship was also proposed.
Collapse
Affiliation(s)
- Youfu Luo
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Honokiol is a small-molecule polyphenol isolated from the genus Magnolia. It is accompanied by other related polyphenols, including magnolol, with which it shares certain biologic properties. Recently, honokiol has been found to have antiangiogenic, antiinflammatory, and antitumor properties in preclinical models, without appreciable toxicity. These findings have increased interest in bringing honokiol to the clinic as a novel chemotherapeutic agent. In addition, mechanistic studies have tried to find the mechanism(s) of action of honokiol, for two major reasons. First, knowledge of the mechanisms of action may assist development of novel synthetic analogues. Second, mechanistic actions of honokiol may lead to rational combinations with conventional chemotherapy or radiation for enhanced response to systemic cancers. In this review, we describe the findings that honokiol has two major mechanisms of action. First, it blocks signaling in tumors with defective p53 function and activated ras by directly blocking the activation of phospholipase D by activated ras. Second, honokiol induces cyclophilin D, thus potentiating the mitochondrial permeability transition pore, and causing death in cells with wild-type p53. Knowledge of the dual activities of honokiol can assist with the development of honokiol derivatives and the design of clinical trials that will maximize the potential benefit of honokiol in the patient setting.
Collapse
Affiliation(s)
- Levi E Fried
- Department of Dermatology, Emory University School of Medicine, Winship Cancer Institute, Atlanta VA Medical Center, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
38
|
Hu J, Chen LJ, Liu L, Chen X, Chen PL, Yang G, Hou WL, Tang MH, Zhang F, Wang XH, Zhao X, Wei YQ. Liposomal honokiol, a potent anti-angiogenesis agent, in combination with radiotherapy produces a synergistic antitumor efficacy without increasing toxicity. Exp Mol Med 2009; 40:617-28. [PMID: 19116447 DOI: 10.3858/emm.2008.40.6.617] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Honokiol is an active compound purified from magnolia that has been shown to induce cell differentiation, apoptosis, and anti-angiogenesis effects, as well as an enhancement in tumor growth delay in combination with chemotherapeutic agents in several mouse xenograft models. Our goal was to investigate the radiosensitization effect of honokiol on lung carcinoma. The radiosensitization effect of liposomal honokiol in Lewis lung carcinoma cells (LL/2) was analyzed using an in vitro clonogenic survival assay. For an in vivo study, Lewis lung carcinoma-bearing C57BL/6 mice were treated with either liposomal honokiol at 25 mg/kg or 5 Gy of single tumor radiation, or a combination of both over 12 days of treatment. The tumor growth delay and the survival time were evaluated. In addition, histological analysis of tumor sections was performed to examine changes by detecting the microvessel density and apoptosis in tumor tissues. In the clonogenic survival assay, LL/2 cells treated with IC(50) Lipo-HNK for 24 h showed a radiation enhancement ratio of 1.9. After 12 days of combination treatment, the tumor volume decreased 78% and produced an anti-tumor activity 1.3-fold greater than a predicted additive effect of honokiol and radiation alone. This combination treatment also caused an 8.7 day delay in tumor growth. The cell cycle distribution and histological analysis demonstrated that liposomal honokiol has an anti-tumor effect via inducing apoptosis and inhibiting angiogenesis. Liposomal honokiol can enhance tumor cell radiosensitivity in vitro and in vivo, indicating that radiotherapy combined with liposomal honokiol can lead to greater anti-tumor efficacy.
Collapse
Affiliation(s)
- Jia Hu
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wen J, Fu AF, Chen LJ, Xie XJ, Yang GL, Chen XC, Wang YS, Li J, Chen P, Tang MH, Shao XM, Lu Y, Zhao X, Wei YQ. Liposomal honokiol inhibits VEGF-D-induced lymphangiogenesis and metastasis in xenograft tumor model. Int J Cancer 2009; 124:2709-18. [PMID: 19219913 DOI: 10.1002/ijc.24244] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Lymph nodes metastasis of tumor could be a crucial early step in the metastatic process. Induction of tumor lymphangiogenesis by vascular endothelial growth factor-D may play an important role in promoting tumor metastasis to regional lymph nodes and these processes can be inhibited by inactivation of the VEGFR-3 signaling pathway. Honokiol has been reported to possess potent antiangiogenesis and antitumor properties in several cell lines and xenograft tumor models. However, its role in tumor-associated lymphangiogenesis and lymphatic metastasis remains unclear. Here, we established lymph node metastasis models by injecting overexpressing VEGF-D Lewis lung carcinoma cells into C57BL/6 mice to explore the effect of honokiol on tumor-associated lymphangiogenesis and related lymph node metastasis. The underlying mechanisms were systematically investigated in vitro and in vivo. In in vivo study, liposomal honokiol significantly inhibited the tumor-associated lymphangiogenesis and metastasis in Lewis lung carcinoma model. A remarkable delay of tumor growth and prolonged life span were also observed. In in vitro study, honokiol inhibited VEGF-D-induced survival, proliferation and tube-formation of both human umbilical vein endothelial cells (HUVECs) and lymphatic vascular endothelial cells (HLECs). Western blotting analysis showed that liposomal honokiol-inhibited Akt and MAPK phosphorylation in 2 endothelial cells, and downregulated expressions of VEGFR-2 of human vascular endothelial cells and VEGFR-3 of lymphatic endothelial cells. Thus, we identified for the first time that honokiol provided therapeutic benefit not only by direct effects on tumor cells and antiangiogenesis but also by inhibiting lymphangiogenesis and metastasis via the VEGFR-3 pathway. The present findings may be of importance to investigate the molecular mechanisms underlying the spread of cancer via the lymphatics and explore the therapeutical strategy of honokiol on antilymphangiogenesis and antimetastasis.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ainbinder D, Protokin R, Chaouat M, Touitou E. Effect of honokiol and 5-FU on non-melanoma skin cancer cells. J Drug Deliv Sci Technol 2009. [DOI: 10.1016/s1773-2247(09)50053-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
41
|
Preparative purification of anti-tumor derivatives of honokiol by high-speed counter-current chromatography. J Chromatogr A 2008; 1178:160-5. [DOI: 10.1016/j.chroma.2007.11.072] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Revised: 11/20/2007] [Accepted: 11/26/2007] [Indexed: 11/20/2022]
|