1
|
Omran MM, Kamal MM, Ammar YA, Abusaif MS, Ismail MMF, Mansour HH. Pharmacological investigation of new niclosamide-based isatin hybrids as antiproliferative, antioxidant, and apoptosis inducers. Sci Rep 2024; 14:19818. [PMID: 39191850 DOI: 10.1038/s41598-024-69250-5] [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: 03/07/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
A group of Niclosamide-linked isatin hybrids (Xo, X1, and X2) was created and examined using IR, 1HNMR, 13C NMR, and mass spectrometry. These hybrids' cytotoxicity, antioxidant, cell cycle analysis, and apoptosis-inducing capabilities were identified. Using the SRB assay, their cytotoxicity against the human HCT-116, MCF-7, and HEPG-2 cancer cell lines, as well as VERO (African Green Monkey Kidney), was evaluated. Compound X1 was the most effective compound. In HCT-116 cells, compound X1 produced cell cycle arrest in the G1 phase, promoted cell death, and induced apoptosis through mitochondrial membrane potential breakdown in comparison to niclosamide and the control. Niclosamide and compound X1 reduced reactive oxygen species generation and modulated the gene expression of BAX, Bcl-2, Bcl-xL, and PAR-4 in comparison to the control. Docking modeling indicated their probable binding modalities with the XIAP BIR2 domain, which selectively binds caspase-3/7, and highlighted their structural drivers of activity for further optimization investigations. Computational in silico modeling of the new hybrids revealed that they presented acceptable physicochemical values as well as drug-like characteristics, which may introduce them as drug-like candidates. The study proved that compound X1 might be a novel candidate for the development of anticancer agents as it presents antiproliferative activity mediated by apoptosis.
Collapse
Affiliation(s)
- Mervat M Omran
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mona M Kamal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Yousry A Ammar
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Moustafa S Abusaif
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Magda M F Ismail
- Department of Medicinal Pharmaceutical Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, 11754, Egypt
| | - Heba H Mansour
- Health Radiation Research Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, P.O. Box 29, Nasr City Cairo, Egypt.
| |
Collapse
|
2
|
Mangla B, Mittal P, Kumar P, Aggarwal G. Multifaceted role of erlotinib in various cancer: nanotechnology intervention, patent landscape, and advancements in clinical trials. Med Oncol 2024; 41:173. [PMID: 38864966 DOI: 10.1007/s12032-024-02414-5] [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: 04/17/2024] [Accepted: 05/23/2024] [Indexed: 06/13/2024]
Abstract
Erlotinib (ELB) is a tyrosine kinase inhibitor that targets the activity of Epidermal Growth Factor Receptor (EGFR) protein found in both healthy and cancerous cells. It binds reversibly to the ATP-binding site of the EGFR tyrosine kinase. ELB was approved by the US Food and Drug Administration (FDA) in 2004 for advanced non-small cell lung cancer (NSCLC) treatment in patients who relapsed after at least one other therapy. It was authorized for use with gemcitabine in 2005 for the treatment of advanced pancreatic cancer. In addition to lung cancer, ELB has shown promising results in the treatment of other cancers, including breast, prostate, colon, pancreatic, cervical, ovarian, and head and neck cancers. However, its limited water solubility, as a BCS class II drug, presents biopharmaceutical problems. Nanoformulations have been developed to overcome these issues, including increased solubility, controlled release, enhanced stability, tumor accumulation, reduced toxicity, and overcoming drug resistance. In older patients, ELB management should involve individualized dosing based on age-related changes in drug metabolism and close monitoring for adverse effects. Regular assessments of renal and hepatic functions are essential. This review provides an overview of ELB's role of ELB in treating various cancers, its associated biopharmaceutical issues, and the latest developments in ELB-related nanotechnology interventions. It also covers ELB patents granted in previous years and the ongoing clinical trials.
Collapse
Affiliation(s)
- Bharti Mangla
- Centre for Advanced Formulation and Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Priya Mittal
- Centre for Advanced Formulation and Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Pankaj Kumar
- Centre for Advanced Formulation and Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Geeta Aggarwal
- Centre for Advanced Formulation and Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| |
Collapse
|
3
|
Kang HW, Kim JH, Lee DE, Lee YS, Kim MJ, Kim HS, Fang S, Lee BE, Lee KJ, Yoo J, Kim HJ, Park JS. Combination therapy of niclosamide with gemcitabine inhibited cell proliferation and apoptosis via Wnt/β-catenin/c-Myc signaling pathway by inducing β-catenin ubiquitination in pancreatic cancer. Cancer Biol Ther 2023; 24:2272334. [PMID: 37917550 PMCID: PMC10623893 DOI: 10.1080/15384047.2023.2272334] [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: 02/22/2022] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a type of cancer with high morbidity and mortality rates worldwide. Owing to a lack of therapeutic options, the overall survival rate of patients with pancreatic cancer is low. Gemcitabine has been mainly used to treat patients with pancreatic cancer, but its efficacy is limited by chemoresistance. Therefore, a novel therapeutic agent for PDAC therapy is urgently needed. An anthelminthic drug, niclosamide, has already been researched in breast, lung, colon, and pancreatic cancer as an anti-cancer purpose by re-positioning its original purpose. However, combination therapy of gemcitabine and niclosamide was not informed yet. Here, we found that niclosamide co-administered with gemcitabine significantly inhibited tumorigenesis of pancreatic cancer compared to gemcitabine alone. Further, combining niclosamide and gemcitabine inhibited cell proliferation and induced apoptosis. Niclosamide induced cell cycle arrest at the G1 phase, and the levels of CDK4/6 and cyclin D1 were lowered after gemcitabine treatment. In addition, the combination of these chemical compounds more effectively increased the binding level of activated β-catenin destruction complex and β-catenin to enable phosphorylation, compared to gemcitabine alone. After phosphorylation, niclosamide - gemcitabine upregulated the ubiquitin level, which caused phosphorylated β-catenin to undergo proteasomal degradation; the combination was more potent than gemcitabine alone. Finally, the combination more effectively suppressed tumor growth in vivo, compared to gemcitabine alone. Altogether, our results indicate that niclosamide synergistically enhances the antitumor effect of gemcitabine in pancreatic cancer, by inducing the degradation of β-catenin with ubiquitination. Therefore, this drug combination can potentially be used in PDAC therapy.
Collapse
Affiliation(s)
- Hyeon Woong Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Ju Hyun Kim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Eun Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yun Sun Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myeong Jin Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Hyung Sun Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - SungSoon Fang
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bo Eun Lee
- CHA Organoid Research Center, School of Medicine, CHA University, Seoul, Republic of Korea
- ORGANOIDSCIENCES, Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyung Jin Lee
- CHA Organoid Research Center, School of Medicine, CHA University, Seoul, Republic of Korea
- ORGANOIDSCIENCES, Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jongman Yoo
- CHA Organoid Research Center, School of Medicine, CHA University, Seoul, Republic of Korea
- ORGANOIDSCIENCES, Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hyo Jung Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Seong Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
4
|
Rahman M, Afzal O, Ullah SNM, Alshahrani MY, Alkhathami AG, Altamimi ASA, Almujri SS, Almalki WH, Shorog EM, Alossaimi MA, Mandal AK, abdulrahman A, Sahoo A. Nanomedicine-Based Drug-Targeting in Breast Cancer: Pharmacokinetics, Clinical Progress, and Challenges. ACS OMEGA 2023; 8:48625-48649. [PMID: 38162753 PMCID: PMC10753706 DOI: 10.1021/acsomega.3c07345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024]
Abstract
Breast cancer (BC) is a malignant neoplasm that begins in the breast tissue. After skin cancer, BC is the second most common type of cancer in women. At the end of 2040, the number of newly diagnosed BC cases is projected to increase by over 40%, reaching approximately 3 million worldwide annually. The hormonal and chemotherapeutic approaches based on conventional formulations have inappropriate therapeutic effects and suboptimal pharmacokinetic responses with nonspecific targeting actions. To overcome such issues, the use of nanomedicines, including liposomes, nanoparticles, micelles, hybrid nanoparticles, etc., has gained wider attention in the treatment of BC. Smaller dimensional nanomedicine (especially 50-200 nm) exhibited improved in vivo effectiveness, such as better tissue penetration and more effective tumor suppression through enhanced retention and permeation, as well as active targeting of the drug. Additionally, nanotechnology, which further extended and developed theranostic nanomedicine by incorporating diagnostic and imaging agents in one platform, has been applied to BC. Furthermore, hybrid and theranostic nanomedicine has also been explored for gene delivery as anticancer therapeutics in BC. Moreover, the nanocarriers' size, shape, surface charge, chemical compositions, and surface area play an important role in the nanocarriers' stability, cellular absorption, cytotoxicity, cellular uptake, and toxicity. Additionally, nanomedicine clinical translation for managing BC remains a slow process. However, a few cases are being used clinically, and their progress with the current challenges is addressed in this Review. Therefore, this Review extensively discusses recent advancements in nanomedicine and its clinical challenges in BC.
Collapse
Affiliation(s)
- Mahfoozur Rahman
- Department
of Pharmaceutical Sciences, Shalom Institute of Health and Allied
Sciences, Sam Higginbottom University of
Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India
| | - Obaid Afzal
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Shehla Nasar Mir
Najib Ullah
- Phyto
Pharmaceuticals Research Lab, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences and Research, Jamia
Hamdard University, Hamdard Nagar, New Delhi, Delhi 110062, India
| | - Mohammad Y. Alshahrani
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 61413, Abha 9088, Saudi Arabia
| | - Ali G. Alkhathami
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 61413, Abha 9088, Saudi Arabia
| | | | - Salem Salman Almujri
- Department
of Pharmacology, College of Pharmacy, King
Khalid University, Asir-Abha 61421, Saudi Arabia
| | - Waleed H Almalki
- Department
of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Eman M. Shorog
- Department
of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Manal A Alossaimi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ashok Kumar Mandal
- Department
of Pharmacology, Faculty of Medicine, University
Malaya, Kuala Lumpur 50603, Malaysia
| | - Alhamyani abdulrahman
- Pharmaceuticals
Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha 65779, Saudi Arabia
| | - Ankit Sahoo
- Department
of Pharmaceutical Sciences, Shalom Institute of Health and Allied
Sciences, Sam Higginbottom University of
Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India
| |
Collapse
|
5
|
Padakanti AP, Pawar SD, Kumar P, Chella N. Development and validation of HPLC method for simultaneous estimation of erlotinib and niclosamide from liposomes optimized by screening design. J Liposome Res 2023; 33:268-282. [PMID: 36594184 DOI: 10.1080/08982104.2022.2162540] [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: 08/20/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 01/04/2023]
Abstract
The emerging drug resistance to the approved first-line drug therapy leads to clinical failure in cancer. Drug repurposing studies lead to the identification of many old drugs to be used for cancer treatment. Combining the repurposed drugs (niclosamide) with first-line therapy agents like erlotinib HCl showed improved efficacy by inhibiting erlotinib HCl acquired resistance. But there is a need to develop a sensitive, accurate, and excellent analytical method and drug delivery system for successfully delivering drug combinations. In the current study, an HPLC method was developed and validated for the simultaneous estimation of niclosamide and erlotinib HCl. The retention time of niclosamide and erlotinib hydrochloride was 6.48 and 7.65 min at 333 nm. The developed method was rapid and sensitive to separating the two drugs with reasonable accuracy, precision, robustness, and ruggedness. A Plackett-Burman (PBD) screening design was used to identify the critical parameters affecting liposomal formulation development using particle size, size distribution, zeta potential, and entrapment efficiency as the response. Lipid concentration, drug concentration, hydration temperature, and media volume were critical parameters affecting the particle size, polydispersity index (PDI), ZP, and %EE of the liposomes. The optimized NCM-ERL liposomes showed the particle size (126.05 ± 2.1), PDI (0.498 ± 0.1), ZP (-16.2 ± 0.3), and %EE of NCM and ERL (50.04 ± 2.8 and 05.42 ± 1.3). In vitro release studies indicated the controlled release of the drugs loaded liposomes (87.06 ± 9.93% and 42.33 ± 0.89% in 24 h).
Collapse
Affiliation(s)
- Amruta Prabhakar Padakanti
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Sila village, Changsari, Assam, India
| | - Sachin Dattaram Pawar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Sila village, Changsari, Assam, India
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Sila village, Changsari, Assam, India
| | - Naveen Chella
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Sila village, Changsari, Assam, India
| |
Collapse
|
6
|
Hsu CM, Chang KC, Chuang TM, Chu ML, Lin PW, Liu HS, Kao SY, Liu YC, Huang CT, Wang MH, Yeh TJ, Gau YC, Du JS, Wang HC, Cho SF, Hsiao CE, Tsai Y, Hsiao SY, Hung LC, Yen CH, Hsiao HH. High G9a Expression in DLBCL and Its Inhibition by Niclosamide to Induce Autophagy as a Therapeutic Approach. Cancers (Basel) 2023; 15:4150. [PMID: 37627178 PMCID: PMC10452841 DOI: 10.3390/cancers15164150] [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: 06/01/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is a malignant lymphoid tumor disease that is characterized by heterogeneity, but current treatment does not benefit all patients, which highlights the need to identify oncogenic genes and appropriate drugs. G9a is a histone methyltransferase that catalyzes histone H3 lysine 9 (H3K9) methylation to regulate gene function and expression in various cancers. METHODS TCGA and GTEx data were analyzed using the GEPIA2 platform. Cell viability under drug treatment was assessed using Alamar Blue reagent; the interaction between G9a and niclosamide was assessed using molecular docking analysis; mRNA and protein expression were quantified in DLBCL cell lines. Finally, G9a expression was quantified in 39 DLBCL patient samples. RESULTS The TCGA database analysis revealed higher G9a mRNA expression in DLBCL compared to normal tissues. Niclosamide inhibited DLBCL cell line proliferation in a time- and dose-dependent manner, reducing G9a expression and increasing p62, BECN1, and LC3 gene expression by autophagy pathway regulation. There was a correlation between G9a expression in DLBCL samples and clinical data, showing that advanced cancer stages exhibited a higher proportion of G9a-expressing cells. CONCLUSION G9a overexpression is associated with tumor progression in DLBCL. Niclosamide effectively inhibits DLBCL growth by reducing G9a expression via the cellular autophagy pathway; therefore, G9a is a potential molecular target for the development of therapeutic strategies for DLBCL.
Collapse
Affiliation(s)
- Chin-Mu Hsu
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
| | - Kung-Chao Chang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Tzer-Ming Chuang
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
| | - Man-Ling Chu
- M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.C.); (P.-W.L.); (H.-S.L.)
| | - Pei-Wen Lin
- M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.C.); (P.-W.L.); (H.-S.L.)
| | - Hsiao-Sheng Liu
- M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.C.); (P.-W.L.); (H.-S.L.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Yu Kao
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Yi-Chang Liu
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Tzu Huang
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Min-Hong Wang
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tsung-Jang Yeh
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
| | - Yuh-Ching Gau
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jeng-Shiun Du
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hui-Ching Wang
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Feng Cho
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chi-En Hsiao
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA;
| | - Yuhsin Tsai
- Graduate Institute of Chinese Medicine, School of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
| | - Samuel Yien Hsiao
- Department of Biology, University of Rutgers-Camden, Camden, NJ 08102, USA;
| | - Li-Chuan Hung
- Long-Term Care and Health Management Department, Cheng Shiu University, Kaohsiung 833, Taiwan;
| | - Chia-Hung Yen
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hui-Hua Hsiao
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-M.H.); (T.-M.C.); (Y.-C.L.); (M.-H.W.); (T.-J.Y.); (Y.-C.G.); (J.-S.D.); (H.-C.W.); (S.-F.C.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
7
|
Kauerová T, Pérez-Pérez MJ, Kollar P. Salicylanilides and Their Anticancer Properties. Int J Mol Sci 2023; 24:ijms24021728. [PMID: 36675241 PMCID: PMC9861143 DOI: 10.3390/ijms24021728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Salicylanilides are pharmacologically active compounds with a wide spectrum of biological effects. Halogenated salicylanilides, which have been used for decades in human and veterinary medicine as anthelmintics, have recently emerged as candidates for drug repurposing in oncology. The most prominent example of salicylanilide anthelmintic, that is intensively studied for its potential anticancer properties, is niclosamide. Nevertheless, recent studies have discovered extensive anticancer potential in a number of other salicylanilides. This potential of their anticancer action is mediated most likely by diverse mechanisms of action such as uncoupling of oxidative phosphorylation, inhibition of protein tyrosine kinase epidermal growth factor receptor, modulation of different signaling pathways as Wnt/β-catenin, mTORC1, STAT3, NF-κB and Notch signaling pathways or induction of B-Raf V600E inhibition. Here we provide a comprehensive overview of the current knowledge about the proposed mechanisms of action of anticancer activity of salicylanilides based on preclinical in vitro and in vivo studies, or structural requirements for such an activity.
Collapse
Affiliation(s)
- Tereza Kauerová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
| | | | - Peter Kollar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
- Correspondence: ; Tel.: +420-541-562-892
| |
Collapse
|
8
|
Shi T, Yu H, Blair RH. Integrated regulatory and metabolic networks of the tumor microenvironment for therapeutic target prioritization. Stat Appl Genet Mol Biol 2023; 22:sagmb-2022-0054. [PMID: 37988745 DOI: 10.1515/sagmb-2022-0054] [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: 11/05/2022] [Accepted: 09/28/2023] [Indexed: 11/23/2023]
Abstract
Translation of genomic discovery, such as single-cell sequencing data, to clinical decisions remains a longstanding bottleneck in the field. Meanwhile, computational systems biological models, such as cellular metabolism models and cell signaling pathways, have emerged as powerful approaches to provide efficient predictions in metabolites and gene expression levels, respectively. However, there has been limited research on the integration between these two models. This work develops a methodology for integrating computational models of probabilistic gene regulatory networks with a constraint-based metabolism model. By using probabilistic reasoning with Bayesian Networks, we aim to predict cell-specific changes under different interventions, which are embedded into the constraint-based models of metabolism. Applications to single-cell sequencing data of glioblastoma brain tumors generate predictions about the effects of pharmaceutical interventions on the regulatory network and downstream metabolisms in different cell types from the tumor microenvironment. The model presents possible insights into treatments that could potentially suppress anaerobic metabolism in malignant cells with minimal impact on other cell types' metabolism. The proposed integrated model can guide therapeutic target prioritization, the formulation of combination therapies, and future drug discovery. This model integration framework is also generalizable to other applications, such as different cell types, organisms, and diseases.
Collapse
Affiliation(s)
- Tiange Shi
- University at Buffalo, Biostatistics, Buffalo, USA
| | - Han Yu
- Roswell Park Comprehensive Cancer Center, Biostatistics and Bioinformatics, Buffalo, USA
| | - Rachael Hageman Blair
- University at Buffalo, Biostatistics, Institute for Artificial Intelligence and Data Science, Buffalo, USA
| |
Collapse
|
9
|
Gargantilla M, Persoons L, Kauerová T, del Río N, Daelemans D, Priego EM, Kollar P, Pérez-Pérez MJ. Hybridization Approach to Identify Salicylanilides as Inhibitors of Tubulin Polymerization and Signal Transducers and Activators of Transcription 3 (STAT3). Pharmaceuticals (Basel) 2022; 15:835. [PMID: 35890135 PMCID: PMC9318074 DOI: 10.3390/ph15070835] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 02/06/2023] Open
Abstract
The superimposition of the X-ray complexes of cyclohexanediones (i.e., TUB015), described by our research group, and nocodazole, within the colchicine binding site of tubulin provided an almost perfect overlap of both ligands. This structural information led us to propose hybrids of TUB015 and nocodazole using a salicylanilide core structure. Interestingly, salicylanilides, such as niclosamide, are well-established signal transducers and activators of transcription (STAT3) inhibitors with anticancer properties. Thus, different compounds with this new scaffold have been synthesized with the aim to identify compounds inhibiting tubulin polymerization and/or STAT3 signaling. As a result, we have identified new salicylanilides (6 and 16) that showed significant antiproliferative activity against a panel of cancer cells. Both compounds were able to reduce the levels of p-STAT3Tyr705 without affecting the total expression of STAT3. While compound 6 inhibited tubulin polymerization and arrested the cell cycle of DU145 cells at G2/M, similar to TUB015, compound 16 showed a more potent effect on inhibiting STAT3 phosphorylation and arrested the cell cycle at G1/G0, similar to niclosamide. In both cases, no toxicity towards PBMC cells was detected. Thus, the salicylanilides described here represent a new class of antiproliferative agents affecting tubulin polymerization and/or STAT3 phosphorylation.
Collapse
Affiliation(s)
- Marta Gargantilla
- Instituto de Quimica Medica (IQM, CSIC) c/Juan de la Cierva 3, 28006 Madrid, Spain; (M.G.); (N.d.R.); (E.-M.P.)
| | - Leentje Persoons
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; (L.P.); (D.D.)
| | - Tereza Kauerová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 42 Brno, Czech Republic;
| | - Natalia del Río
- Instituto de Quimica Medica (IQM, CSIC) c/Juan de la Cierva 3, 28006 Madrid, Spain; (M.G.); (N.d.R.); (E.-M.P.)
| | - Dirk Daelemans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; (L.P.); (D.D.)
| | - Eva-María Priego
- Instituto de Quimica Medica (IQM, CSIC) c/Juan de la Cierva 3, 28006 Madrid, Spain; (M.G.); (N.d.R.); (E.-M.P.)
| | - Peter Kollar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 42 Brno, Czech Republic;
| | - María-Jesús Pérez-Pérez
- Instituto de Quimica Medica (IQM, CSIC) c/Juan de la Cierva 3, 28006 Madrid, Spain; (M.G.); (N.d.R.); (E.-M.P.)
| |
Collapse
|
10
|
Singh S, Weiss A, Goodman J, Fisk M, Kulkarni S, Lu I, Gray J, Smith R, Sommer M, Cheriyan J. Niclosamide-A promising treatment for COVID-19. Br J Pharmacol 2022; 179:3250-3267. [PMID: 35348204 PMCID: PMC9111792 DOI: 10.1111/bph.15843] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/09/2022] [Accepted: 02/23/2022] [Indexed: 12/15/2022] Open
Abstract
Vaccines have reduced the transmission and severity of COVID-19, but there remains a paucity of efficacious treatment for drug-resistant strains and more susceptible individuals, particularly those who mount a suboptimal vaccine response, either due to underlying health conditions or concomitant therapies. Repurposing existing drugs is a timely, safe and scientifically robust method for treating pandemics, such as COVID-19. Here, we review the pharmacology and scientific rationale for repurposing niclosamide, an anti-helminth already in human use as a treatment for COVID-19. In addition, its potent antiviral activity, niclosamide has shown pleiotropic anti-inflammatory, antibacterial, bronchodilatory and anticancer effects in numerous preclinical and early clinical studies. The advantages and rationale for nebulized and intranasal formulations of niclosamide, which target the site of the primary infection in COVID-19, are reviewed. Finally, we give an overview of ongoing clinical trials investigating niclosamide as a promising candidate against SARS-CoV-2.
Collapse
Affiliation(s)
- Shivani Singh
- Division of Pulmonary and Critical Care MedicineNYU School of MedicineNew YorkNew YorkUSA
| | - Anne Weiss
- Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKongens LyngbyDenmark
- UNION Therapeutics Research ServicesHellerupDenmark
| | - James Goodman
- Department of MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Marie Fisk
- Department of MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Spoorthy Kulkarni
- Department of MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Ing Lu
- Department of MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Joanna Gray
- Department of MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Rona Smith
- Department of MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
- Cambridge Clinical Trials UnitCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Morten Sommer
- Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKongens LyngbyDenmark
- UNION TherapeuticsHellerupDenmark
| | - Joseph Cheriyan
- Department of MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
- Cambridge Clinical Trials UnitCambridge University Hospitals NHS Foundation TrustCambridgeUK
| |
Collapse
|
11
|
Valdez L, Cheng B, Gonzalez D, Rodriguez R, Campano P, Tsin A, Fang X. Combined treatment with niclosamide and camptothecin enhances anticancer effect in U87 MG human glioblastoma cells. Oncotarget 2022; 13:642-658. [PMID: 35548329 PMCID: PMC9084225 DOI: 10.18632/oncotarget.28227] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Laura Valdez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- These authors contributed equally to this work
| | - Benxu Cheng
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- These authors contributed equally to this work
| | - Daniela Gonzalez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Reanna Rodriguez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Paola Campano
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Andrew Tsin
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Xiaoqian Fang
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
12
|
Pistone M, Racaniello GF, Arduino I, Laquintana V, Lopalco A, Cutrignelli A, Rizzi R, Franco M, Lopedota A, Denora N. Direct cyclodextrin-based powder extrusion 3D printing for one-step production of the BCS class II model drug niclosamide. Drug Deliv Transl Res 2022; 12:1895-1910. [PMID: 35138629 PMCID: PMC9242976 DOI: 10.1007/s13346-022-01124-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 01/02/2023]
Abstract
Niclosamide (NCS) is a drug that has been used as an anthelmintic and anti-parasitic drug for about 40 years. Recently, some studies have highlighted its potential in treating various tumors, allowing a repositioning of this drug. Despite its potential, NCS is a Biopharmaceutical Classification System (BCS) Class II drug and is consequently characterized by low aqueous solubility, poor dissolution rate and reduced bioavailability, which limits its applicability. In this work, we utilize a very novel technique, direct powder extrusion (DPE) 3D printing, which overcomes the limitations of previously used techniques (fused deposition modelling, FDM) to achieve direct extrusion of powder mixtures consisting of NCS, hydroxypropyl methylcellulose (HPMC, Affinisol 15 LV), hydroxypropyl-β-cyclodextrin (HP-β-CD) and polyethylene glycol (PEG) 6000. For the first time, direct printing of powder blends containing HP-β-CD was conducted. For all tablets, in vitro dissolution studies showed sustained drug release over 48 h, but for tablets containing HP-β-CD, the release was faster. Solid-state characterization studies showed that during extrusion, the drug lost its crystal structure and was evenly distributed within the polymer matrix. All printed tablets have exhibited good mechanical and physical features and a stability of the drug content for up to 3 months. This innovative printing technique has demonstrated the possibility to produce personalized pharmaceutical forms directly from powders, avoiding the use of filament used by FDM.
Collapse
Affiliation(s)
- Monica Pistone
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy
| | | | - Ilaria Arduino
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy
| | - Valentino Laquintana
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy
| | - Antonio Lopalco
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy
| | - Annalisa Cutrignelli
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy
| | - Rosanna Rizzi
- Institute of Crystallography-CNR, Amendola St. 122/o, 70126, Bari, Italy
| | - Massimo Franco
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy
| | - Angela Lopedota
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy.
| | - Nunzio Denora
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Orabona St. 4, 70125, Bari, Italy.
| |
Collapse
|
13
|
Trefoil Factor 3 Inhibits Thyroid Cancer Cell Progression Related to IL-6/JAK/STAT3 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2130229. [PMID: 34567204 PMCID: PMC8457945 DOI: 10.1155/2021/2130229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/27/2021] [Indexed: 11/18/2022]
Abstract
Objectives Abnormal expression of trefoil factor 3 (TFF3) in breast, stomach, and colon tumors may be related to the occurrence of tumors, suggesting its role in angiogenesis. In this study, the aim was to explore the role of TFF3 in thyroid cancer. Methods TFF3 expression analysis was performed via GEPIA and RT-PCR. To explore the effects of TFF3 on thyroid cancer cell motility, cell function assays were performed. Furthermore, GSEA pathway analysis and western blot were used to explore the mechanism by which TFF3 represses the progression of thyroid cancer cells. Results Here, we showed that low expression level of TFF3 in thyroid cancer is related to thyroid cancer nodal metastasis. The patients with low TFF3 expression showed worse disease-free survival than those with high level of TFF3. Underexpressed TFF3 increased cell motility and inhibited cell apoptosis. We found that the levels of IL-6, p-JAK2/JAK2, and pSTAT3/STAT3 were inhibited in the pcDNA-TFF3 group compared to the pcDNA-NC group and these factors were upregulated in the si-TFF3 group compared to the si-NC group in BCPAP and TPC-1 cells. Conclusion TFF3 inhibits thyroid cancer cell progression related to IL-6/JAK/STAT3 signaling pathway.
Collapse
|
14
|
Recurrence in Oral Premalignancy: Clinicopathologic and Immunohistochemical Analysis. Diagnostics (Basel) 2021; 11:diagnostics11050872. [PMID: 34066207 PMCID: PMC8151734 DOI: 10.3390/diagnostics11050872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/13/2023] Open
Abstract
Oral leukoplakia (OL) has a propensity for recurrence and malignant transformation (MT). Herein, we evaluate sociodemographic, clinical, microscopic and immunohistochemical parameters as predictive factors for OL recurrence, also comparing primary lesions (PLs) with recurrences. Thirty-three patients with OL, completely removed either by excisional biopsy or by laser ablation following incisional biopsy, were studied. Selected molecules associated with the STAT3 oncogenic pathway, including pSTAT3, Bcl-xL, survivin, cyclin D1 and Ki-67, were further analyzed. A total of 135 OL lesions, including 97 PLs and 38 recurrences, were included. Out of 97 PLs, 31 recurred at least once and none of them underwent MT, during a mean follow-up time of 48.3 months. There was no statistically significant difference among the various parameters in recurrent vs. non-recurrent PLs, although recurrence was most frequent in non-homogeneous lesions (p = 0.087) and dysplastic lesions recurred at a higher percentage compared to hyperplastic lesions (34.5% vs. 15.4%). Lower levels of Bcl-xL and survivin were identified as significant risk factors for OL recurrence. Recurrences, although smaller and more frequently homogeneous and non-dysplastic compared to their corresponding PLs, exhibited increased immunohistochemical expression of oncogenic molecules, especially pSTAT3 and Bcl-xL. Our results suggest that parameters associated with recurrence may differ from those that affect the risk of progression to malignancy and support OL management protocols favoring excision and close monitoring of all lesions.
Collapse
|
15
|
Wang G, Gaikwad H, McCarthy MK, Gonzalez-Juarrero M, Li Y, Armstrong M, Reisdorph N, Morrison TE, Simberg D. Lipid nanoparticle formulation of niclosamide (nano NCM) effectively inhibits SARS-CoV-2 replication in vitro. PRECISION NANOMEDICINE 2021; 4:724-737. [PMID: 34676370 PMCID: PMC8528232 DOI: 10.33218/001c.18813] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As exemplified by the COVID-19 pandemic, highly infective respiratory viruses can spread rapidly in the population because of lack of effective approaches to control viral replication and spread. Niclosamide (NCM) is an old anthelminthic drug (World Health Organization essential medicine list) with pleiotropic pharmacological activities. Several recent publications demonstrated that NCM has broad antiviral activities and potently inhibits viral replication, including replication of SARS-CoV-2, SARS-CoV, and dengue viruses. Unfortunately, NCM is almost completely insoluble in water, which limits its clinical use. We developed a cost-effective lipid nanoparticle formulation of NCM (nano NCM) using only FDA-approved excipient and demonstrated potency against SARS-CoV-2 infection in cells (Vero E6 and ACE2-expressing lung epithelium cells).
Collapse
Affiliation(s)
- Guankui Wang
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Mary K McCarthy
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Mercedes Gonzalez-Juarrero
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80521
| | - Yue Li
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Thomas E Morrison
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| |
Collapse
|
16
|
Fujii A, Masuda T, Iwata M, Tobo T, Wakiyama H, Koike K, Kosai K, Nakano T, Kuramitsu S, Kitagawa A, Sato K, Kouyama Y, Shimizu D, Matsumoto Y, Utsunomiya T, Ohtsuka T, Yamanishi Y, Nakamura M, Mimori K. The novel driver gene ASAP2 is a potential druggable target in pancreatic cancer. Cancer Sci 2021; 112:1655-1668. [PMID: 33605496 PMCID: PMC8019229 DOI: 10.1111/cas.14858] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022] Open
Abstract
Targeting mutated oncogenes is an effective approach for treating cancer. The 4 main driver genes of pancreatic ductal adenocarcinoma (PDAC) are KRAS, TP53, CDKN2A, and SMAD4, collectively called the "big 4" of PDAC, however they remain challenging therapeutic targets. In this study, ArfGAP with SH3 domain, ankyrin repeat and PH domain 2 (ASAP2), one of the ArfGAP family, was identified as a novel driver gene in PDAC. Clinical analysis with PDAC datasets showed that ASAP2 was overexpressed in PDAC cells based on increased DNA copy numbers, and high ASAP2 expression contributed to a poor prognosis in PDAC. The biological roles of ASAP2 were investigated using ASAP2-knockout PDAC cells generated with CRISPR-Cas9 technology or transfected PDAC cells. In vitro and in vivo analyses showed that ASAP2 promoted tumor growth by facilitating cell cycle progression through phosphorylation of epidermal growth factor receptor (EGFR). A repositioned drug targeting the ASAP2 pathway was identified using a bioinformatics approach. The gene perturbation correlation method showed that niclosamide, an antiparasitic drug, suppressed PDAC growth by inhibition of ASAP2 expression. These data show that ASAP2 is a novel druggable driver gene that activates the EGFR signaling pathway. Furthermore, niclosamide was identified as a repositioned therapeutic agent for PDAC possibly targeting ASAP2.
Collapse
Affiliation(s)
- Atsushi Fujii
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
- Department of Surgery and OncologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takaaki Masuda
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Michio Iwata
- Department of Bioscience and BioinformaticsFaculty of Computer Science and Systems EngineeringKyushu Institute of TechnologyFukuokaJapan
| | - Taro Tobo
- Department of Clinical Laboratory MedicineKyushu University Beppu HospitalOitaJapan
| | | | - Kensuke Koike
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Keisuke Kosai
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Takafumi Nakano
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | | | | | - Kuniaki Sato
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Yuta Kouyama
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | - Dai Shimizu
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| | | | | | - Takao Ohtsuka
- Department of Digestive Surgery, Breast and Thyroid SurgeryKagoshima UniversityKagoshimaJapan
| | - Yoshihiro Yamanishi
- Department of Bioscience and BioinformaticsFaculty of Computer Science and Systems EngineeringKyushu Institute of TechnologyFukuokaJapan
| | - Masafumi Nakamura
- Department of Surgery and OncologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koshi Mimori
- Department of SurgeryKyushu University Beppu HospitalOitaJapan
| |
Collapse
|
17
|
Arshad U, Pertinez H, Box H, Tatham L, Rajoli RKR, Curley P, Neary M, Sharp J, Liptrott NJ, Valentijn A, David C, Rannard SP, O’Neill PM, Aljayyoussi G, Pennington SH, Ward SA, Hill A, Back DJ, Khoo SH, Bray PG, Biagini GA, Owen A. Prioritization of Anti-SARS-Cov-2 Drug Repurposing Opportunities Based on Plasma and Target Site Concentrations Derived from their Established Human Pharmacokinetics. Clin Pharmacol Ther 2020; 108:775-790. [PMID: 32438446 PMCID: PMC7280633 DOI: 10.1002/cpt.1909] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
There is a rapidly expanding literature on the in vitro antiviral activity of drugs that may be repurposed for therapy or chemoprophylaxis against severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). However, this has not been accompanied by a comprehensive evaluation of the target plasma and lung concentrations of these drugs following approved dosing in humans. Accordingly, concentration 90% (EC90 ) values recalculated from in vitro anti-SARS-CoV-2 activity data was expressed as a ratio to the achievable maximum plasma concentration (Cmax ) at an approved dose in humans (Cmax /EC90 ratio). Only 14 of the 56 analyzed drugs achieved a Cmax /EC90 ratio above 1. A more in-depth assessment demonstrated that only nitazoxanide, nelfinavir, tipranavir (ritonavir-boosted), and sulfadoxine achieved plasma concentrations above their reported anti-SARS-CoV-2 activity across their entire approved dosing interval. An unbound lung to plasma tissue partition coefficient (Kp Ulung ) was also simulated to derive a lung Cmax /half-maximal effective concentration (EC50 ) as a better indicator of potential human efficacy. Hydroxychloroquine, chloroquine, mefloquine, atazanavir (ritonavir-boosted), tipranavir (ritonavir-boosted), ivermectin, azithromycin, and lopinavir (ritonavir-boosted) were all predicted to achieve lung concentrations over 10-fold higher than their reported EC50 . Nitazoxanide and sulfadoxine also exceeded their reported EC50 by 7.8-fold and 1.5-fold in lung, respectively. This analysis may be used to select potential candidates for further clinical testing, while deprioritizing compounds unlikely to attain target concentrations for antiviral activity. Future studies should focus on EC90 values and discuss findings in the context of achievable exposures in humans, especially within target compartments, such as the lungs, in order to maximize the potential for success of proposed human clinical trials.
Collapse
Affiliation(s)
- Usman Arshad
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Henry Pertinez
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Helen Box
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Lee Tatham
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Rajith K. R. Rajoli
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Paul Curley
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Megan Neary
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Joanne Sharp
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Neill J. Liptrott
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Anthony Valentijn
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Christopher David
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | | | | | - Ghaith Aljayyoussi
- Department of Tropical Disease BiologyLiverpool School of Tropical MedicineCentre for Drugs and DiagnosticsLiverpoolUK
| | - Shaun H. Pennington
- Department of Tropical Disease BiologyLiverpool School of Tropical MedicineCentre for Drugs and DiagnosticsLiverpoolUK
| | - Stephen A. Ward
- Department of Tropical Disease BiologyLiverpool School of Tropical MedicineCentre for Drugs and DiagnosticsLiverpoolUK
| | - Andrew Hill
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - David J. Back
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | - Saye H. Khoo
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| | | | - Giancarlo A. Biagini
- Department of Tropical Disease BiologyLiverpool School of Tropical MedicineCentre for Drugs and DiagnosticsLiverpoolUK
| | - Andrew Owen
- Department of Molecular and Clinical PharmacologyMaterials Innovation FactoryUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
18
|
Weisberg E, Parent A, Yang PL, Sattler M, Liu Q, Liu Q, Wang J, Meng C, Buhrlage SJ, Gray N, Griffin JD. Repurposing of Kinase Inhibitors for Treatment of COVID-19. Pharm Res 2020; 37:167. [PMID: 32778962 PMCID: PMC7417114 DOI: 10.1007/s11095-020-02851-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022]
Abstract
The outbreak of COVID-19, the pandemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spurred an intense search for treatments by the scientific community. In the absence of a vaccine, the goal is to target the viral life cycle and alleviate the lung-damaging symptoms of infection, which can be life-threatening. There are numerous protein kinases associated with these processes that can be inhibited by FDA-approved drugs, the repurposing of which presents an alluring option as they have been thoroughly vetted for safety and are more readily available for treatment of patients and testing in clinical trials. Here, we characterize more than 30 approved kinase inhibitors in terms of their antiviral potential, due to their measured potency against key kinases required for viral entry, metabolism, or reproduction. We also highlight inhibitors with potential to reverse pulmonary insufficiency because of their anti-inflammatory activity, cytokine suppression, or antifibrotic activity. Certain agents are projected to be dual-purpose drugs in terms of antiviral activity and alleviation of disease symptoms, however drug combination is also an option for inhibitors with optimal pharmacokinetic properties that allow safe and efficacious co-administration with other drugs, such as antiviral agents, IL-6 blocking agents, or other kinase inhibitors.
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Alexander Parent
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Priscilla L Yang
- Department of Cancer Cell Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Qingsong Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Qingwang Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jinhua Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Chengcheng Meng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara J Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Nathanael Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - James D Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Wei W, Liu H, Yuan J, Yao Y. Targeting Wnt/β‐catenin by anthelmintic drug niclosamide overcomes paclitaxel resistance in esophageal cancer. Fundam Clin Pharmacol 2020; 35:165-173. [PMID: 32579788 DOI: 10.1111/fcp.12583] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 06/13/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Wei Wei
- Department of Oncology Xiangyang Central Hospital Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| | - Hongfang Liu
- Department of Oncology Xiangyang Central Hospital Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| | - Jia Yuan
- Department of Oncology Xiangyang Central Hospital Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| | - Yang Yao
- Department of Oncology Xiangyang Central Hospital Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| |
Collapse
|
20
|
SS Pindiprolu SK, Krishnamurthy PT, Ghanta VR, Chintamaneni PK. Phenyl boronic acid-modified lipid nanocarriers of niclosamide for targeting triple-negative breast cancer. Nanomedicine (Lond) 2020; 15:1551-1565. [DOI: 10.2217/nnm-2020-0003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To study the active targeting efficacy of phenylboronic acid-modified niclosamide solid lipid nanoparticles (PBA-Niclo-SLN) in triple-negative breast cancer (TNBC). Materials & methods: PBA-Niclo-SLNs were formulated by an emulsification-solvent evaporation method using PBA-associated stearylamine (PBSA) as lipid. The drug uptake and the anticancer propensity of PBA-Niclo-SLN were studied in TNBC (MDA-MB231) cells and tumor-bearing mice. Results: PBA-Niclo-SLN formulation resulted in greater antitumor efficacy by inducing G0/G1 cell cycle arrest and apoptosis. Besides, PBA-Niclo-SLN effectively inhibited STAT3, CD44+/CD24- TNBC stem cell subpopulation, epithelial–mesenchymal transition markers. Besides, PBA-Niclo-SLN selectively accumulated at the tumor site with more significant tumor regression and improved the survivability in TNBC tumor-bearing mice. Conclusion: PBA-Niclo-SLN formulation would be an effective strategy to eradicate TNBC cells (breast cancer stem cells and nonbreast cancer stem cells) efficiently.
Collapse
Affiliation(s)
- Sai Kiran SS Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, 643001, Tamil Nadu, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, 643001, Tamil Nadu, India
| | - Venkata Rao Ghanta
- Synthetic Organic Chemistry Division, GVK Biosciences Private Limited, IDA Nacharam, Hyderabad, 500076, Telangana, India
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, 643001, Tamil Nadu, India
| |
Collapse
|
21
|
Ring-Substituted 1-Hydroxynaphthalene-2-Carboxanilides Inhibit Proliferation and Trigger Mitochondria-Mediated Apoptosis. Int J Mol Sci 2020; 21:ijms21103416. [PMID: 32408543 PMCID: PMC7279329 DOI: 10.3390/ijms21103416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/05/2020] [Accepted: 05/10/2020] [Indexed: 12/13/2022] Open
Abstract
Ring-substituted 1-hydroxynaphthalene-2-carboxanilides were previously investigated for their antimycobacterial properties. In our study, we have shown their antiproliferative and cell death-inducing effects in cancer cell lines. Cell proliferation and viability were assessed by WST-1 assay and a dye exclusion test, respectively. Cell cycle distribution, phosphatidylserine externalization, levels of reactive oxygen or nitrogen species (RONS), mitochondrial membrane depolarization, and release of cytochrome c were estimated by flow cytometry. Levels of regulatory proteins were determined by Western blotting. Our data suggest that the ability to inhibit the proliferation of THP-1 or MCF-7 cells might be referred to meta- or para-substituted derivatives with electron-withdrawing groups -F, -Br, or -CF3 at anilide moiety. This effect was accompanied by accumulation of cells in G1 phase. Compound 10 also induced apoptosis in THP-1 cells in association with a loss of mitochondrial membrane potential and production of mitochondrial superoxide. Our study provides a new insight into the action of salicylanilide derivatives, hydroxynaphthalene carboxamides, in cancer cells. Thus, their structure merits further investigation as a model moiety of new small-molecule compounds with potential anticancer properties.
Collapse
|
22
|
Jensen KV, Hao X, Aman A, Luchman HA, Weiss S. EGFR blockade in GBM brain tumor stem cells synergizes with JAK2/STAT3 pathway inhibition to abrogate compensatory mechanisms in vitro and in vivo. Neurooncol Adv 2020; 2:vdaa020. [PMID: 32226941 PMCID: PMC7086303 DOI: 10.1093/noajnl/vdaa020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background The EGFR pathway is frequently mutated in glioblastoma (GBM). However, to date, EGFR therapies have not demonstrated efficacy in clinical trials. Poor brain penetration of conventional inhibitors, lack of patient stratification for EGFR status, and mechanisms of resistance are likely responsible for the failure of EGFR-targeted therapy. We aimed to address these elements in a large panel of molecularly diverse patient-derived GBM brain tumor stem cells (BTSCs). Methods In vitro growth inhibition and on-target efficacy of afatinib, pacritinib, or a combination were assessed by cell viability, neurosphere formation, cytotoxicity, limiting dilution assays, and western blotting. In vivo efficacy was assessed with mass spectrometry, immunohistochemistry, magnetic resonance imaging, and intracranial xenograft models. Results We show that afatinib and pacritinib decreased BTSC growth and sphere-forming capacity in vitro. Combinations of the 2 drugs were synergistic and abrogated the activation of STAT3 signaling observed upon EGFR inhibition in vitro and in vivo. We further demonstrate that the brain-penetrant EGFR inhibitor, afatinib, improved survival in EGFRvIII mt orthotopic xenograft models. However, upregulation of the oncogenic STAT3 signaling pathway was observed following afatinib treatment. Combined inhibition with 2 clinically relevant drugs, afatinib and pacritinib, synergistically decreased BTSC viability and abrogated this compensatory mechanism of resistance to EGFR inhibition. A significant decrease in tumor burden in vivo was observed with the combinatorial treatment. Conclusions These data demonstrate that brain-penetrant combinatorial therapies targeting the EGFR and STAT3 signaling pathways hold therapeutic promise for GBM.
Collapse
Affiliation(s)
- Katharine V Jensen
- Hotchkiss Brain Institute and Arnie Charbonneau Cancer Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Xiaoguang Hao
- Hotchkiss Brain Institute and Arnie Charbonneau Cancer Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - H Artee Luchman
- Hotchkiss Brain Institute and Arnie Charbonneau Cancer Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Hotchkiss Brain Institute and Arnie Charbonneau Cancer Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Lee MC, Chen YK, Hsu YJ, Lin BR. Niclosamide inhibits the cell proliferation and enhances the responsiveness of esophageal cancer cells to chemotherapeutic agents. Oncol Rep 2019; 43:549-561. [PMID: 31894334 PMCID: PMC6967135 DOI: 10.3892/or.2019.7449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Niclosamide is an FDA-approved anthelmintic drug, and may elicit antineoplastic effects through direct STAT3 inhibition, which has been revealed in numerous human cancer cells. Chemotherapy is the standard treatment for advanced esophageal cancers, but also causes severe systemic side effects. The present study represents the first study evaluating the anticancer efficacy of niclosamide in esophageal cancers. Through western blot assay, it was demonstrated that niclosamide suppressed the STAT3 signaling pathway in esophageal adenocarcinoma cells (BE3) and esophageal squamous cell carcinoma cells (CE48T and CE81T). In addition, niclosamide inhibited cell proliferation as determined by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay and soft agar colony forming assay, and induced cell apoptosis as determined by Annexin V and PI staining. The induction of p21 and G1 arrest of the cell cycle also was revealed in niclosamide-treated CE81T cells by qPCR and flow cytometric assays, respectively. Furthermore, in the combination analysis of niclosamide and chemotherapeutic agents by MTS assay, low IC50 values were detected in cells co-treated with niclosamide, with the exception of cisplatin-treated CE81T cells. To confirm the results using an apoptosis assay, the apoptotic enhancement of niclosamide was only demonstrated in CE48T cells co-treated with 5-FU, cisplatin, or paclitaxel, and in BE3 cells co-treated with paclitaxel, but not in CE81T cells. These findings indicate a future clinical application of niclosamide in esophageal cancers.
Collapse
Affiliation(s)
- Ming-Cheng Lee
- Department of Research and Development, DrSignal BioTechnology Ltd., New Taipei City 23143, Taiwan, R.O.C
| | - Yin-Kai Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan, R.O.C
| | - Yih-Jen Hsu
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei 10051, Taiwan, R.O.C
| | - Bor-Ru Lin
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei 10051, Taiwan, R.O.C
| |
Collapse
|
24
|
Ashrafizadeh M, Ahmadi Z, Kotla NG, Afshar EG, Samarghandian S, Mandegary A, Pardakhty A, Mohammadinejad R, Sethi G. Nanoparticles Targeting STATs in Cancer Therapy. Cells 2019; 8:E1158. [PMID: 31569687 PMCID: PMC6829305 DOI: 10.3390/cells8101158] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past decades, an increase in the incidence rate of cancer has been witnessed. Although many efforts have been made to manage and treat this life threatening condition, it is still one of the leading causes of death worldwide. Therefore, scientists have attempted to target molecular signaling pathways involved in cancer initiation and metastasis. It has been shown that signal transducers and activator of transcription (STAT) contributes to the progression of cancer cells. This important signaling pathway is associated with a number of biological processes including cell cycle, differentiation, proliferation and apoptosis. It appears that dysregulation of the STAT signaling pathway promotes the migration, viability and malignancy of various tumor cells. Hence, there have been many attempts to target the STAT signaling pathway. However, it seems that currently applied therapeutics may not be able to effectively modulate the STAT signaling pathway and suffer from a variety of drawbacks such as low bioavailability and lack of specific tumor targeting. In the present review, we demonstrate how nanocarriers can be successfully applied for encapsulation of STAT modulators in cancer therapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran.
| | - Zahra Ahmadi
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar 6451741117, Iran.
| | - Niranjan G Kotla
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Newcastle, Galway H91 W2TY, Ireland.
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran.
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran.
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran.
| | - Abbas Pardakhty
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran.
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| |
Collapse
|
25
|
Hatamipour M, Jaafari MR, Momtazi-Borojeni AA, Ramezani M, Sahebkar A. Evaluation of the Anti-Tumor Activity of Niclosamide Nanoliposomes Against Colon Carcinoma. Curr Mol Pharmacol 2019; 13:245-250. [PMID: 31433764 DOI: 10.2174/1874467212666190821142721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Niclosamide is an established anti-helminthic drug, which has recently been shown to inhibit the growth of various cancer cells. To exploit the potential anti-tumor activity of this drug for systemic use, the problem of low aqueous solubility should be addressed. The present study tested the in vivo anti-tumor effects of a recently developed nanoliposomal preparation of niclosamide in an experimental model of colon carcinoma. METHODS The cytotoxicity of nanoliposomal niclosamide on CT26 colon carcinoma cells was evaluated using the MTT test. Inhibition of tumor growth was investigated in BALB/c mice bearing CT26 colon carcinoma cells. The animals were randomly divided into 4 groups including: 1) untreated control, 2) liposomal doxorubicin (15 mg/kg; single intravenous dose), 3) liposomal niclosamide (1 mg/kg/twice a week; intravenously for 4 weeks), and 4) free niclosamide (1 mg/kg/twice a week; intravenously for 4 weeks). To study therapeutic efficacy, tumor size and survival were monitored in 2-day intervals for 40 days. RESULTS In vitro results indicated that nanoliposomal and free niclosamide could exert cytotoxic effects with IC50 values of 4.5 and 2.5 μM, respectively. According to in vivo studies, nanoliposomal niclosamide showed a higher growth inhibitory activity against CT26 colon carcinoma cells compared with free niclosamide as revealed by delayed tumor growth and prolongation of survival. CONCLUSION Nnaoliposomal encapsulation enhanced anti-tumor properties of niclosamide in an experimental model of colon carcinoma.
Collapse
Affiliation(s)
- Mahdi Hatamipour
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences,
Mashhad, Iran
| | - Mahmoud R Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences,
Mashhad, Iran,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical
Sciences, Mashhad, Iran
| | | | - Mahin Ramezani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences,
Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical
Sciences, Mashhad, Iran,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Wang CF, Zhang HC, Feng XM, Song XM, Wu YN. Knockdown of MSI1 inhibits the proliferation of human oral squamous cell carcinoma by inactivating STAT3 signaling. Int J Mol Med 2019; 44:115-124. [PMID: 31059073 PMCID: PMC6559311 DOI: 10.3892/ijmm.2019.4181] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
Musashi RNA-binding protein 1 (MSI1) is highly expressed in several types of cancer; however, its role in oral squamous cell carcinoma (OSCC) remains unknown. The purpose of this study was to investigate the probable mechanism underlying the involvement of MSI1 in OSCC. The results demonstrated that MSI1 was upregulated in OSCC tissues, but not in adjacent healthy tissues. MSI1 silencing resulted in decreased cell proliferative, invasive and migrative capacity. In addition, MSI1 silencing led to cell cycle arrest at the S phase, downregulation of c-Myc and cyclin D1, and upregulation of p21 and p27 levels. Additional studies demonstrated that MSI1 suppression inhibited the activation of signal transducer and activator of transcription 3 (STAT3) signaling. Accordingly, the findings of the present study suggested that MSI1 silencing can suppress OSCC cell proliferation and progression, in part by inhibiting the activation of the c-Myc/STAT3 pathway.
Collapse
Affiliation(s)
- Chen-Fei Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Hong-Chuang Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Xin-Mei Feng
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 216000, P.R. China
| | - Xiao-Meng Song
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Yu-Nong Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
27
|
Fan X, Xu J, Files M, Cirillo JD, Endsley JJ, Zhou J, Endsley MA. Dual activity of niclosamide to suppress replication of integrated HIV-1 and Mycobacterium tuberculosis (Beijing). Tuberculosis (Edinb) 2019; 116S:S28-S33. [PMID: 31080089 PMCID: PMC7106448 DOI: 10.1016/j.tube.2019.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 11/24/2022]
Abstract
The human immunodeficiency virus (HIV) pandemic is driving the re-emergence of tuberculosis (TB) as a global health threat, both by increasing the susceptibility of HIV-infected people to infection with Mycobacterium tuberculosis (Mtb), and increasing the rate of emergence of drug-resistant Mtb. There are several other clinical challenges for treatment of co-infected patients including: expense, pill burden, toxicity, and malabsorption that further necessitate the search for new drugs that may be effective against both pathogens simultaneously. The anti-helminthic niclosamide has been shown to have activity against a laboratory strain of Mtb in liquid culture while bacteriostatic activity against non-replicating M. abscessus was also recently described. Here we extend these findings to further demonstrate that niclosamide inhibits mycobacterial growth in infected human macrophages and mediates potent bacteriostatic activity against the virulent Mtb Beijing strain. Importantly, we provide the first evidence that niclosamide inhibits HIV replication in human macrophages and Jurkat T cells through post-integration effects on pro-virus transcription. The dual antiviral and anti-mycobacterial activity was further observed in an in vitro model of HIV and Mtb co-infection using human primary monocyte-derived macrophages. These results support further investigation of niclosamide and derivatives as anti-retroviral/anti-mycobacterial agents that may reduce clinical challenges associated with multi-drug regimens and drug resistance.
Collapse
Affiliation(s)
- XiuZhen Fan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Jimin Xu
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Megan Files
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Jeffrey D Cirillo
- Department of Microbial Pathogenesis and Immunology, Center for Airborne Pathogen Research and Tuberculosis Imaging, Texas A&M Health Sciences Center, College Station, TX, USA.
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Mark A Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
28
|
Han Z, Li Q, Wang Y, Wang L, Li X, Ge N, Wang Y, Guo C. Niclosamide Induces Cell Cycle Arrest in G1 Phase in Head and Neck Squamous Cell Carcinoma Through Let-7d/CDC34 Axis. Front Pharmacol 2019; 9:1544. [PMID: 30687101 PMCID: PMC6333743 DOI: 10.3389/fphar.2018.01544] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/18/2018] [Indexed: 01/11/2023] Open
Abstract
Niclosamide is a traditional anti-tapeworm drug that exhibits potent anti-cancer activity. Our previous study showed that niclosamide induces cell cycle arrest in G1 phase. Nevertheless, the underlying mechanism remains unknown. The following study investigated the molecular mechanism through which niclosamide induced G1 arrest in head and neck squamous cell carcinoma (HNSCC) cell lines. The effect of niclosamide on human HNSCC cell line WSU-HN6 and CNE-2Z were analyzed using IncuCyte ZOOMTM assay, flow cytometry (FCM), real-time PCR and western blot. Luciferase assay was conducted to demonstrate the interaction between let-7d (a let-7 family member which functions as a tumor suppressor by regulating cell cycle) and 3′UTR of CDC34 mRNA. Xenografts tumor model was established to evaluate the niclosamide treatment efficacy in vivo. Briefly, an exposure to niclosamide treatment led to an increased let-7d expression and a decreased expression of cell cycle regulator CDC34, finally leading to G1 phase arrest. Moreover, an overexpression of let-7d induced G1 phase arrest and downregulated CDC34, while the knockdown of let-7d partially rescued the niclosamide-induced G1 phase arrest. Luciferase assay confirmed the direct inhibition of CDC34 through the targeting of let-7d. Furthermore, niclosamide markedly inhibited the xenografts growth through up-regulation of let-7d and down-regulation of CDC34. To sum up, our findings suggest that niclosamide induces cell cycle arrest in G1 phase in HNSCC through let-7d/CDC34 axis, which enriches the anti-cancer mechanism of niclosamide.
Collapse
Affiliation(s)
- Zewen Han
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qingxiang Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yifei Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaoxu Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Na Ge
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yixiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Chuanbin Guo
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
29
|
Pindiprolu SKSS, Chintamaneni PK, Krishnamurthy PT, Ratna Sree Ganapathineedi K. Formulation-optimization of solid lipid nanocarrier system of STAT3 inhibitor to improve its activity in triple negative breast cancer cells. Drug Dev Ind Pharm 2018; 45:304-313. [DOI: 10.1080/03639045.2018.1539496] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Sai Kiran S. S. Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy, Ooty (Constituent College), JSS Academy of Higher Education and Research, Mysuru, India
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy, Ooty (Constituent College), JSS Academy of Higher Education and Research, Mysuru, India
| | - Praveen T. Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, Ooty (Constituent College), JSS Academy of Higher Education and Research, Mysuru, India
| | - Kinnera Ratna Sree Ganapathineedi
- Department of Pharmacology, JSS College of Pharmacy, Ooty (Constituent College), JSS Academy of Higher Education and Research, Mysuru, India
| |
Collapse
|
30
|
Wang C, Zhou X, Xu H, Shi X, Zhao J, Yang M, Zhang L, Jin X, Hu Y, Li X, Xiao X, Liao M. Niclosamide Inhibits Cell Growth and Enhances Drug Sensitivity of Hepatocellular Carcinoma Cells via STAT3 Signaling Pathway. J Cancer 2018; 9:4150-4155. [PMID: 30519314 PMCID: PMC6277621 DOI: 10.7150/jca.26948] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/07/2018] [Indexed: 01/19/2023] Open
Abstract
Aims: Hepatocellular carcinoma (HCC) is the sixth most common malignant tumor worldwide, with a high mortality rate at advanced stages. In this study, we investigated the effect of niclosamide on cell growth and drug sensitivity in human HCC and elucidated the underlying mechanism. Methods: Three human HCC cell lines (HepG2, QGY-7703 and SMMC-7721) were used to evaluate the effect of niclosamide. Cell proliferation was measured by MTT assay and colony formation assay. Assessment of apoptosis was evaluated by flow cytometry and Hoechst staining. The mRNA and protein levels were analyzed by real-time PCR and western blot, respectively. Results: Niclosamide suppressed cell viability, inhibited clone formation, and induced cell apoptosis in HCC cells dose- and time-dependently. Furthermore, niclosamide synergized with cisplatin to promote the apoptosis of HCC cells. With niclosamide treatment, phospho-STAT3 (Y705) was inactivated and the downstream antiapoptotic proteins Mcl-1 and survivin were downregulated at both mRNA and protein levels in HCC cells. Conclusion: Niclosamide has effective function in anti-HCC and may be a single or combined drug treatment for HCC and acts via the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Chengzhi Wang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.,Department of Nephrology, Xiangya Hospital,Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xiaoqing Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Hongjuan Xu
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xiaqing Shi
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jinfeng Zhao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Manyi Yang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Lihua Zhang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xin Jin
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Yu Hu
- Center for Experimental Medical Research, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P.R. China
| | - Xia Li
- Department of Nephrology, Xiangya Hospital,Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital,Central South University, Changsha, Hunan, 410008, P.R. China
| | - Mingmei Liao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| |
Collapse
|
31
|
Involvement of Up-Regulation of DR5 Expression and Down-Regulation of c-FLIP in Niclosamide-Mediated TRAIL Sensitization in Human Renal Carcinoma Caki Cells. Molecules 2018; 23:molecules23092264. [PMID: 30189637 PMCID: PMC6225471 DOI: 10.3390/molecules23092264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 01/01/2023] Open
Abstract
Niclosamide is used to treat intestinal parasite infections, as being an anthelmintic drug. Recently, several papers suggest the niclosamide inhibits multiple signaling pathways, which are highly activated and mutated in cancer. Here, niclosamide was evaluated for identifying strategies to overcome tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resistance. Although niclosamide (100–200 nM) alone did not bring about cell death, combinations of niclosamide and TRAIL led to apoptotic cell death in carcinoma cells, but not in normal cells. Niclosamide markedly increased DR5 protein levels, including cell-surface DR5, and decreased c-FLIP protein levels. Down-regulation of DR5 by specific small interfering RNA (siRNA) and ectopic expression of c-FLIP markedly blocked niclosamide plus TRAIL-induced apoptosis. Our findings provide that niclosamide could overcome resistance to TRAIL through up-regulating DR5 on the cell surface and down-regulating c-FLIP in cancer cells. Taken together, niclosamide may be an attractive candidate to overcome TRAIL resistance.
Collapse
|
32
|
Kadri H, Lambourne OA, Mehellou Y. Niclosamide, a Drug with Many (Re)purposes. ChemMedChem 2018; 13:1088-1091. [PMID: 29603892 PMCID: PMC7162286 DOI: 10.1002/cmdc.201800100] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/29/2018] [Indexed: 12/13/2022]
Abstract
Niclosamide is an anthelmintic drug that has been used for over 50 years mainly to treat tapeworm infections. However, with the increase in drug repurposing initiatives, niclosamide has emerged as a true hit in many screens against various diseases. Indeed, from being an anthelmintic drug, it has now shown potential in treating Parkinson's disease, diabetes, viral and microbial infections, as well as various cancers. Such diverse pharmacological activities are a result of niclosamide's ability to uncouple mitochondrial phosphorylation and modulate a selection of signaling pathways, such as Wnt/β-catenin, mTOR and JAK/STAT3, which are implicated in many diseases. In this highlight, we discuss the plethora of diseases that niclosamide has shown promise in treating.
Collapse
Affiliation(s)
- Hachemi Kadri
- Cardiff School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Olivia A Lambourne
- Cardiff School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Youcef Mehellou
- Cardiff School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| |
Collapse
|
33
|
Burock S, Daum S, Keilholz U, Neumann K, Walther W, Stein U. Phase II trial to investigate the safety and efficacy of orally applied niclosamide in patients with metachronous or sychronous metastases of a colorectal cancer progressing after therapy: the NIKOLO trial. BMC Cancer 2018; 18:297. [PMID: 29544454 PMCID: PMC5856000 DOI: 10.1186/s12885-018-4197-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 03/07/2018] [Indexed: 02/08/2023] Open
Affiliation(s)
- Susen Burock
- Charité Comprehensive Cancer Center, Invalidenstraße 80, 10117, Berlin, Germany.
| | - Severin Daum
- Department of Medicine I, Gastroenterology, Rheumatology and Infectious Diseases, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Invalidenstraße 80, 10117, Berlin, Germany.,German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Konrad Neumann
- Charité Comprehensive Cancer Center, Invalidenstraße 80, 10117, Berlin, Germany.,Department for Biostatistics and Clinical Epidemiology, Charité Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
34
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK. Pharmacological targets of breast cancer stem cells: a review. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:463-479. [PMID: 29476201 DOI: 10.1007/s00210-018-1479-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023]
Abstract
Breast cancers contain small population of tumor-initiating cells called breast cancer stem cells (BCSCs), which are spared even after chemotherapy. Recently, BCSCs are implicated to be a cause of metastasis, tumor relapse, and therapy resistance in breast cancer. BCSCs have unique molecular mechanisms, which can be targeted to eliminate them. These include surface biomarkers, proteins involved in self-renewal pathways, drug efflux transporters, apoptotic/antiapoptotic proteins, autophagy, metabolism, and microenvironment regulation. The complex molecular mechanisms behind the survival of BCSCs and pharmacological targets for elimination of BCSCs are described in this review.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India.
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| |
Collapse
|
35
|
Li J, Li H, Zhan D, Xiang M, Yang J, Zuo Y, Yu Y, Zhou H, Jiang D, Luo H, Chen Z, Yu Z, Xu Z. Niclosamide sensitizes nasopharyngeal carcinoma to radiation by downregulating Ku70/80 expression. J Cancer 2018; 9:736-744. [PMID: 29556331 PMCID: PMC5858495 DOI: 10.7150/jca.20963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 12/13/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate whether niclosamide could sensitize the nasopharyngeal carcinoma cells to radiation and further explore the underlying mechanisms. CCK-8 assay was used to determine the effect of niclosamide on the proliferation of NPC cells. Colony formation assay was used to evaluate the radiosensitizing effect of niclosamide on NPC cells. Flow cytometry analysis was used to determine the apoptosis of NPC cells induced by niclosamide. Immunofluorescent staining was used to detect the formation of γ-H2AX foci and the localization of Ku70/80 proteins in NPC cells. Real-time PCR quantification analysis was used to examine the level of Ku70/80 mRNA. DNA damage repair-related proteins were detected by western blot analysis. Our results showed that niclosamide markedly suppressed the proliferation of NPC cells. Niclosamide pretreatment followed by irradiation reduced the colony forming ability of NPC cells. Niclosamide in combination with irradiation significantly increased the apoptotic rate of NPC cells. Niclosamide significantly reduced the transcriptional level of K70/80 but not the translocation of Ku70/80 protein induced by irradiation. In conclusion, our study demonstrated that niclosamide could inhibit the growth of NPC cells and sensitize the NPC cells to radiation via suppressing the transcription of Ku70/80.
Collapse
Affiliation(s)
- Jingjing Li
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Haiwen Li
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Dechao Zhan
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Mei Xiang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Jun Yang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Yufang Zuo
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Yin Yu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Hechao Zhou
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Danxian Jiang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Haiqing Luo
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Zihong Chen
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Zhonghua Yu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Zumin Xu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| |
Collapse
|
36
|
Zuo Y, Yang D, Yu Y, Xiang M, Li H, Yang J, Li J, Jiang D, Zhou H, Xu Z, Yu Z. Niclosamide enhances the cytotoxic effect of cisplatin in cisplatin-resistant human lung cancer cells via suppression of lung resistance-related protein and c-myc. Mol Med Rep 2017; 17:3497-3502. [PMID: 29257330 PMCID: PMC5802146 DOI: 10.3892/mmr.2017.8301] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/11/2017] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is a leading cause of cancer-associated mortality worldwide. The cisplatin (DDP)-based chemotherapy remains the foundation of treatment for the majority of patients affected by advanced non-small cell lung cancer (NSCLC). However, DDP-resistance limits the clinical utility of this drug in patients with advanced NSCLC. The aim of the present study was to investigate the inhibitory effect of niclosamide on human lung cancer cell growth and to investigate the possible underlying mechanism. The effects of niclosamide on the proliferation of human lung adenocarcinoma (A549) and DDP-resistant (CR) human lung adenocarcinoma (A549/DDP) cells were examined by Cell Counting kit-8 assay. The impact of niclosamide on the apoptosis of A549/DDP cells was detected by Annexin V-fluorescein isothiocyanate/propidium iodide assay. The expression levels of cisplatin-resistant-associated molecules (lung resistance-related protein and c-myc) following niclosamide treatment in A549/DDP cells were evaluated by western blot analysis. The results indicated that niclosamide in combination with DDP demonstrated a synergistic effect in A549/DDP cells and directly induced apoptosis, which may be associated with caspase-3 activation. Furthermore, niclosamide decreased the expression level of c-myc protein, which may influence DDP sensitivity of A549/DDP cells. Thus, the present study indicates that niclosamide combined with DDP exerts a synergistic effect in cisplatin-resistant lung cancer cells and may present as a promising drug candidate in lung cancer therapy.
Collapse
Affiliation(s)
- Yufang Zuo
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Dongyan Yang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yin Yu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Mei Xiang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Haiwen Li
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jun Yang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jingjing Li
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Danxian Jiang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Hechao Zhou
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Zumin Xu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Zhonghua Yu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| |
Collapse
|
37
|
Misra SK, De A, Pan D. Targeted Delivery of STAT-3 Modulator to Breast Cancer Stem-Like Cells Downregulates a Series of Stemness Genes. Mol Cancer Ther 2017; 17:119-129. [PMID: 29138265 DOI: 10.1158/1535-7163.mct-17-0070] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 09/05/2017] [Accepted: 10/26/2017] [Indexed: 11/16/2022]
Abstract
Cancer stem cells are known to be controlled by pathways that are dormant in normal adult cells, for example, PTEN, which is a negative regulator of transcription factor STAT3. STAT3 regulates genes that are involved in stem cell self-renewal and thus represents a novel therapeutic target of enormous clinical significance. Studies on breast cancer stem cells (BCSC) have been also significantly correlated with STATs. We describe here for the first time a novel strategy to selectively target CSCs and to induce downregulation of STAT3 downstream target genes reducing expression of series of "stem-ness genes" in treated tumors. In vitro and in vivo experiments were performed to evaluate functional activity with gene and protein expression studies. The results of the study indicate that this targeted delivery approach deactivates STAT3 causing a reduction of CD44+/CD24- CSC populations with aptly tracked gene and protein regulations of "stemness" characteristics. Mol Cancer Ther; 17(1); 119-29. ©2017 AACR.
Collapse
Affiliation(s)
- Santosh K Misra
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, Illinois.,Department of Materials Science and Engineering, University of Illinois at Urbana Champaign, Urbana, Illinois.,Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois
| | - Arun De
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, Illinois.,Department of Materials Science and Engineering, University of Illinois at Urbana Champaign, Urbana, Illinois.,Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, Illinois. .,Department of Materials Science and Engineering, University of Illinois at Urbana Champaign, Urbana, Illinois.,Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois
| |
Collapse
|
38
|
Li X, Ding R, Han Z, Ma Z, Wang Y. Targeting of cell cycle and let-7a/STAT3 pathway by niclosamide inhibits proliferation, migration and invasion in oral squamous cell carcinoma cells. Biomed Pharmacother 2017; 96:434-442. [PMID: 29031202 DOI: 10.1016/j.biopha.2017.09.149] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/24/2017] [Accepted: 09/13/2017] [Indexed: 02/08/2023] Open
Abstract
The low median survival rate of oral squamous cell carcinoma (OSCC) is associated with chemotherapeutic resistance. Niclosamide is an oral anti-helminthic drug, its anti-cancer effect has been reported in recent years. However, the effect of niclosamide on OSCC remains largely unknown. In this study, we, for the first time, investigated the underlying mechanisms from cell cycle arrest and let-7a/STAT3 axis through CCK-8, cell cycle, apoptosis, wound healing, Transwell invasion, generation of stable cell line, real-time PCR, and western blot assays using two OSCC cell lines WSU-HN6 and Tca83. We showed that niclosamide could inhibit OSCC cells proliferation through causing cell cycle arrest in G1 phase and promoting apoptosis, while the cell cycle-related proteins MCM2, MCM7, CDK2 and CDK4 were downregulated and the apoptosis-related proteins p53 and cleaved caspase-3 were upregulated. Furthermore, niclosamide could inhibit migration and invasion of OSCC through upregulation of let-7a expression and downregulation of p-STAT3 expression. What is more, we established the stably expressing let-7a cell line (HN6-let-7a). Like niclosamide, HN6-let-7a could decrease the ability of the cell migration, invasion as well as the expression of p-STAT3. Collectively, our study finds the new mechanisms that niclosamide inhibits OSCC proliferation through causing cell cycle arrest in G1 phase via downregulation of the above cell cycle-related genes; promotes OSCC apoptosis through upregulation of pro-apoptotic genes; decreases migration and invasion of OSCC by let-7a/STAT3 axis, thus providing a preferred therapeutic candidate for OSCC in future.
Collapse
Affiliation(s)
- Xiaoxu Li
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China; Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China, China
| | - Ruiyu Ding
- Department of VIP Service, Peking University School and Hospital of Stomatology, Beijing, China
| | - Zewen Han
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China, China
| | - Zeyun Ma
- Department of VIP Service, Peking University School and Hospital of Stomatology, Beijing, China.
| | - Yixiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China; Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China, China.
| |
Collapse
|
39
|
Cheng B, Morales LD, Zhang Y, Mito S, Tsin A. Niclosamide induces protein ubiquitination and inhibits multiple pro-survival signaling pathways in the human glioblastoma U-87 MG cell line. PLoS One 2017; 12:e0184324. [PMID: 28877265 PMCID: PMC5587337 DOI: 10.1371/journal.pone.0184324] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/22/2017] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma is the most common and lethal malignant primary brain tumor for which the development of efficacious chemotherapeutic agents remains an urgent need. The anti-helminthic drug niclosamide, which has long been in use to treat tapeworm infections, has recently attracted renewed interest due to its apparent anticancer effects in a variety of in vitro and in vivo cancer models. However, the mechanism(s) of action remains to be elucidated. In the present study, we found that niclosamide induced cell toxicity in human glioblastoma cells corresponding with increased protein ubiquitination, ER stress and autophagy. In addition, niclosamide treatment led to down-regulation of Wnt/β-catenin, PI3K/AKT, MAPK/ERK, and STAT3 pro-survival signal transduction pathways to further reduce U-87 MG cell viability. Taken together, these results provide new insights into the glioblastoma suppressive capabilities of niclosamide, showing that niclosamide can target multiple major cell signaling pathways simultaneously to effectively promote cell death in U-87 MG cells. Niclosamide constitutes a new prospect for a therapeutic treatment against human glioblastoma.
Collapse
Affiliation(s)
- Benxu Cheng
- Department of Biomedical Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
- * E-mail:
| | - Liza Doreen Morales
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| | - Yonghong Zhang
- Department of Chemistry, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| | - Shizue Mito
- Department of Chemistry, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| | - Andrew Tsin
- Department of Biomedical Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| |
Collapse
|
40
|
Arkhipov IA, Sadov KM, Limova YV, Sadova AK, Varlamova AI, Khalikov SS, Dushkin AV, Chistyachenko YS. The efficacy of the supramolecular complexes of niclosamide obtained by mechanochemical technology and targeted delivery against cestode infection of animals. Vet Parasitol 2017; 246:25-29. [PMID: 28969776 DOI: 10.1016/j.vetpar.2017.08.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 10/18/2022]
Abstract
Niclosamide is an anthelmintic that is widely used to treat cestode infection of animals. The efficacy of the supramolecular complexes of niclosamide obtained by mechanochemical technology and targeted delivery was studied in hymenolepiosis of mice and monieziosis of sheep. The efficacy of new substances of niclosamide with polyvinylpyrrolidone polymer in different ratios (1:10; 1:5; 1:2) was determined by the results of helminthological necropsy of the small intestine of sheep and mice. Pre-treatment eggs per gram (EPG) were not significantly different (P>0.1) among groups. The controlled test was used to evaluate the efficacy. A high efficacy (>95% efficacy) of the supramolecular complexes of niclosamide with PVP (SCoNwPVP) was shown in different ratios (1:10; 1:5 and 1:2) at a dose of 20mg/kg of body weight at oral administration against Hymenolepis nana in mice and Moniezia expansa in sheep. Whereas the basic drug - substance of niclosamide was effective at a dose of 100mg/kg of b/w. No adverse effects of the drugs on animal health were detected during the study.
Collapse
Affiliation(s)
- Ivan A Arkhipov
- All-Russian Scientific Research Institute of Fundamental and Applied Parasitology of Animals and Plants named after K.I. Skryabin, Bolshaya Cheremushkinskaya street 28, 117218 Moscow, Russia
| | - Konstantin M Sadov
- Scientific Research Veterinary Station of the Russian Academy of Agricultural Sciences, Magnitogorskaya street 8, 446013 Samara, Russia
| | - Yulia V Limova
- Scientific Research Veterinary Station of the Russian Academy of Agricultural Sciences, Magnitogorskaya street 8, 446013 Samara, Russia
| | - Alexandra K Sadova
- Scientific Research Veterinary Station of the Russian Academy of Agricultural Sciences, Magnitogorskaya street 8, 446013 Samara, Russia
| | - Anastasiya I Varlamova
- All-Russian Scientific Research Institute of Fundamental and Applied Parasitology of Animals and Plants named after K.I. Skryabin, Bolshaya Cheremushkinskaya street 28, 117218 Moscow, Russia.
| | - Salavat S Khalikov
- A.N. Nesmeyanov Institute of Organoelement Compounds of the Russian Academy of Sciences, Vavilova street 28, 119991 Moscow, Russia
| | - Alexandr V Dushkin
- Institute of Solid State Chemistry and Mechanochemistry of the Siberian Branch of the Russian Academy of Sciences, 630128, Novosibirsk, Kutateladze str., 18, Russia
| | - Yulia S Chistyachenko
- Institute of Solid State Chemistry and Mechanochemistry of the Siberian Branch of the Russian Academy of Sciences, 630128, Novosibirsk, Kutateladze str., 18, Russia
| |
Collapse
|
41
|
Xiang M, Chen Z, Yang D, Li H, Zuo Y, Li J, Zhang W, Zhou H, Jiang D, Xu Z, Yu Z. Niclosamide enhances the antitumor effects of radiation by inhibiting the hypoxia-inducible factor-1α/vascular endothelial growth factor signaling pathway in human lung cancer cells. Oncol Lett 2017; 14:1933-1938. [PMID: 28781637 PMCID: PMC5530112 DOI: 10.3892/ol.2017.6372] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/04/2017] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is one of the leading causes of cancer-associated mortality, worldwide. The overall survival rate remains low, but progress has been made in improving the diagnosis and treatment of lung cancer over the past decades. Niclosamide, a salicylanilide derivative used for the treatment of tapeworm infections, is safe, well tolerated, inexpensive and readily available. Previous studies have identified niclosamide as a potential anticancer agent. The present study demonstrated that niclosamide enhanced the effect of irradiation by inhibiting the hypoxia-inducible factor-1α/vascular endothelial growth factor signaling pathway. These findings suggest that niclosamide may be a promising candidate for clinical evaluation as part of a combined regimen for the treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
- Mei Xiang
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Zihong Chen
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Donghong Yang
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Haiwen Li
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yufang Zuo
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jingjing Li
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Wendian Zhang
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Hechao Zhou
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Danxian Jiang
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Zumin Xu
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Zhonghua Yu
- The Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| |
Collapse
|
42
|
Shi L, Zheng H, Hu W, Zhou B, Dai X, Zhang Y, Liu Z, Wu X, Zhao C, Liang G. Niclosamide inhibition of STAT3 synergizes with erlotinib in human colon cancer. Onco Targets Ther 2017; 10:1767-1776. [PMID: 28367059 PMCID: PMC5370071 DOI: 10.2147/ott.s129449] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Niclosamide, an anthelmintic drug approved by the US Food and Drug Administration against cestodes, is used to treat tapeworm infection. In this study, we show that niclosamide can potentially inhibit signal transducer and activator of transcription 3 (STAT3) in colon cancer cell lines. Combined inhibition of epidermal growth factor receptor and STAT3 by erlotinib and niclosamide synergistically induces apoptosis and antiproliferation in colon cancer cell lines. Our findings suggest that erlotinib and niclosamide combination provides an effective therapeutic approach to improving the prognosis of colon cancer.
Collapse
Affiliation(s)
- Lingyi Shi
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Hailun Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Wanle Hu
- Department of Coloproctology, The Second Affiliated Hospital and Yuying Children's Hospital
| | - Bin Zhou
- Department of Coloproctology, The Second Affiliated Hospital and Yuying Children's Hospital
| | - Xuanxuan Dai
- Department of Oncological Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yi Zhang
- Department of Oncological Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhiguo Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Xiaoping Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Chengguang Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| |
Collapse
|
43
|
Lin CK, Bai MY, Hu TM, Wang YC, Chao TK, Weng SJ, Huang RL, Su PH, Lai HC. Preclinical evaluation of a nanoformulated antihelminthic, niclosamide, in ovarian cancer. Oncotarget 2016; 7:8993-9006. [PMID: 26848771 PMCID: PMC4891020 DOI: 10.18632/oncotarget.7113] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer treatment remains a challenge and targeting cancer stem cells presents a promising strategy. Niclosamide is an “old” antihelminthic drug that uncouples mitochondria of intestinal parasites. Although recent studies demonstrated that niclosamide could be a potential anticancer agent, its poor water solubility needs to be overcome before further preclinical and clinical investigations can be conducted. Therefore, we evaluated a novel nanosuspension of niclosamide (nano-NI) for its effect against ovarian cancer. Nano-NI effectively inhibited the growth of ovarian cancer cells in which it induced a metabolic shift to glycolysis at a concentration of less than 3 μM in vitro and suppressed tumor growth without obvious toxicity at an oral dose of 100 mg/kg in vivo. In a pharmacokinetic study after oral administration, nano-NI showed rapid absorption (reaching the maximum plasma concentration within 5 min) and improved the bioavailability (the estimated bioavailability for oral nano-NI was 25%). In conclusion, nano-NI has the potential to be a new treatment modality for ovarian cancer and, therefore, further clinical trials are warranted.
Collapse
Affiliation(s)
- Chi-Kang Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Tri-Service General Hospital, Taipei, Taiwan
| | - Meng-Yi Bai
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Teh-Min Hu
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Chi Wang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Tri-Service General Hospital, Taipei, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shao-Ju Weng
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Rui-Lan Huang
- Department of Obstetrics and Gynecology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hsuan Su
- Department of Obstetrics and Gynecology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hung-Cheng Lai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
44
|
Morin F, Kavian N, Nicco C, Cerles O, Chéreau C, Batteux F. Niclosamide Prevents Systemic Sclerosis in a Reactive Oxygen Species–Induced Mouse Model. THE JOURNAL OF IMMUNOLOGY 2016; 197:3018-3028. [DOI: 10.4049/jimmunol.1502482] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 08/14/2016] [Indexed: 12/27/2022]
|
45
|
Jung J, Gleave Parson M, Kraft JD, Lyda L, Kobe B, Davis C, Robinson J, Peña MMO, Robinson CM. Elevated interleukin-27 levels in human neonatal macrophages regulate indoleamine dioxygenase in a STAT-1 and STAT-3-dependent manner. Immunology 2016; 149:35-47. [PMID: 27238498 PMCID: PMC4981608 DOI: 10.1111/imm.12625] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 12/12/2022] Open
Abstract
Microbial infections are a major cause of infant mortality as a result of limitations in immune defences. Interleukin-27 (IL-27) is a heterodimeric cytokine produced primarily by leucocytes and is immunosuppressive toward lymphocytes and leucocytes. Our laboratory demonstrated that human neonatal macrophages express IL-27 more abundantly than adult macrophages. Similarly in mice, IL-27 expression is elevated early in life and maintained through infancy. To determine IL-27-regulated mechanisms that may limit immunity, we evaluated the expression of a number of genes in response to this cytokine in primary human neonatal macrophages. Indoleamine 2,3-dioxygenase (IDO) gene expression was increased dose-responsively by IL-27. We have previously demonstrated inhibition of T-cell proliferation and cytokine production by neonatal macrophage-generated IL-27, and IDO is often implicated in this negative regulation. An increase in IDO protein was demonstrated by immunofluorescence microscopy and was consistent with increased enzyme activity following treatment with IL-27. Inclusion of a soluble receptor to neutralize endogenous IL-27, decreased IDO expression and activity compared with untreated macrophages. In response to IL-27, neonatal macrophages phosphorylate signal transdcuer and activator of transcription 1 (STAT-1) and STAT-3. Both transcription factors are recruited to the IDO regulatory region. STAT-3 dominates during steady-state regulation by lower levels of endogenous IL-27 production. A shift to enhanced STAT-1 recruitment occurs during increased levels of exogenously supplied IL-27. These data suggest an interesting interplay of STAT-1 and STAT-3 to regulate IDO activity and immunosuppression in response to different levels of IL-27 in the microenvironment of the immune response that may further our understanding of this interesting cytokine.
Collapse
Affiliation(s)
- Joo‐Yong Jung
- Department of BiologyBriar Cliff UniversitySioux CityIAUSA
| | - Madeline Gleave Parson
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| | - Jennifer D. Kraft
- Department of Pathology Microbiology and ImmunologyUniversity of South Carolina School of MedicineColumbiaSCUSA
| | - Logan Lyda
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| | - Brianna Kobe
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| | - Celestia Davis
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Jembber Robinson
- Department of Pathology Microbiology and ImmunologyUniversity of South Carolina School of MedicineColumbiaSCUSA
| | - Maria Marjorette O. Peña
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Cory M. Robinson
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| |
Collapse
|
46
|
Prather GR, MacLean JA, Shi M, Boadu DK, Paquet M, Hayashi K. Niclosamide As a Potential Nonsteroidal Therapy for Endometriosis That Preserves Reproductive Function in an Experimental Mouse Model. Biol Reprod 2016; 95:76. [PMID: 27535961 PMCID: PMC5333938 DOI: 10.1095/biolreprod.116.140236] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/16/2016] [Indexed: 01/05/2023] Open
Abstract
Endometriosis causes severe chronic pelvic pain and infertility. Because the standard medication and surgical treatments of endometriosis show high recurrence of symptoms, it is necessary to improve current treatment options. In the initial study, we examined whether niclosamide can be a useful drug for endometriosis in a preclinical setting. Endometriotic implants were induced using an established mouse model involving transimplantation of mouse endometrial fragments to the peritoneal wall of recipient mice. When the recipient mice were treated with niclosamide for 3 weeks, niclosamide reduced the size of endometriotic implants with inhibition of cell proliferation, and inflammatory signaling including RELA (NFKB) and STAT3 activation, but did not alter expression of steroid hormone receptors. To identify genes whose expression is regulated by niclosamide in endometriotic implants, RNA-sequencing was performed, and several genes downregulated by niclosamide were related to inflammatory responses, WNT and MAPK signaling. In a second study designed to assess whether niclosamide affects reproductive function, the recipient mice started receiving niclosamide after the induction of endometriosis. Then, the recipient mice were mated with wild type males, and treatments continued until the pups were born. Niclosamide treated recipient mice became pregnant and produced normal size and number of pups. These results suggest that niclosamide could be an effective therapeutic drug, and acts as an inhibitor of inflammatory signaling without disrupting normal reproductive function.
Collapse
Affiliation(s)
- Genna R. Prather
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - James A. MacLean
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Mingxin Shi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Daniel K. Boadu
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Marilène Paquet
- Departement de Pathologie et de Microbiologie, Université de Montreal, St-Hyacinthe, Quebec, Canada
| | - Kanako Hayashi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
- Correspondence: Kanako Hayashi, Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Dr., Carbondale, IL 62901. E-mail:
| |
Collapse
|
47
|
Morin F, Kavian N, Nicco C, Cerles O, Chéreau C, Batteux F. Improvement of Sclerodermatous Graft-Versus-Host Disease in Mice by Niclosamide. J Invest Dermatol 2016; 136:2158-2167. [PMID: 27424318 DOI: 10.1016/j.jid.2016.06.624] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/22/2016] [Accepted: 06/27/2016] [Indexed: 01/19/2023]
Abstract
Sclerodermatous graft-versus-host disease, a frequent complication of allogeneic hematopoietic stem cell graft, shares many features with systemic sclerosis, such as production of autoantibodies and fibrosis of skin and inner organs. Recent reports on the implication of signal transducer and activator of transcription 3 and of Wnt/β-catenin in fibrosis have prompted us to investigate the effects of the inhibition of both signaling pathways in a mouse model of sclerodermatous graft-versus-host disease, using niclosamide, an anthelmintic drug, with a well-defined safety profile. Sclerodermatous graft-versus-host disease was induced in BALB/c mice by B10.D2 bone marrow and spleen cell transplantation. Mice were treated every other day, 5 days a week, for 5 weeks by niclosamide. Clinical and biological features were studied 42 days after transplantation. Niclosamide reversed clinical symptoms including alopecia, vasculitis, and diarrhea and prevented fibrosis of the skin and visceral organs. Beneficial immunological effects were also observed: niclosamide decreased the production of effector memory CD4 and CD8 T cells, T-cell infiltration of the skin and visceral organs, and decreased productions of IL-4 and IL-13, and autoimmune B-cell activation. The improvement provided by niclosamide in the mouse model of sclerodermatous graft-versus-host disease provides a rationale for the evaluation of niclosamide in the management of patients affected by systemic fibrotic disease.
Collapse
Affiliation(s)
- Florence Morin
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes Sorbonne Paris Cité, Paris, France; Laboratoire d'Immunologie biologique, Hôpital Cochin, Paris, France
| | - Niloufar Kavian
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes Sorbonne Paris Cité, Paris, France; Laboratoire d'Immunologie biologique, Hôpital Cochin, Paris, France
| | - Carole Nicco
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Olivier Cerles
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Christiane Chéreau
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Frédéric Batteux
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes Sorbonne Paris Cité, Paris, France; Laboratoire d'Immunologie biologique, Hôpital Cochin, Paris, France.
| |
Collapse
|
48
|
S100A4 in Cancer Metastasis: Wnt Signaling-Driven Interventions for Metastasis Restriction. Cancers (Basel) 2016; 8:cancers8060059. [PMID: 27331819 PMCID: PMC4931624 DOI: 10.3390/cancers8060059] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/27/2016] [Accepted: 06/09/2016] [Indexed: 02/07/2023] Open
Abstract
The aberrant activity of Wnt signaling is an early step in the transformation of normal intestinal cells to malignant tissue, leading to more aggressive tumors, and eventually metastases. In colorectal cancer (CRC), metastasis accounts for about 90% of patient deaths, representing the most lethal event during the course of the disease and is directly linked to patient survival, critically limiting successful therapy. This review focuses on our studies of the metastasis-inducing gene S100A4, which we identified as transcriptional target of β-catenin. S100A4 increased migration and invasion in vitro and metastasis in mice. In patient CRC samples, high S100A4 levels predict metastasis and reduced patient survival. Our results link pathways important for tumor progression and metastasis: the Wnt signaling pathway and S100A4, which regulates motility and invasiveness. S100A4 suppression by interdicting Wnt signaling has potential for therapeutic intervention. As proof of principle, we applied S100A4 shRNA systemically and prevented metastasis in mice. Furthermore, we identified small molecule inhibitors from high-throughput screens of pharmacologically active compounds employing an S100A4 promoter-driven reporter. Best hits act, as least in part, via intervening in the Wnt pathway and restricted metastasis in mouse models. We currently translate our findings on restricting S100A4-driven metastasis into clinical practice. The repositioned FDA-approved drug niclosamide, targeting Wnt signaling, is being tested in a prospective phase II clinical trial for treatment of CRC patients. Our assay for circulating S100A4 transcripts in patient blood is used to monitor treatment success.
Collapse
|
49
|
Moskaleva EY, Perevozchikova VG, Zhirnik AS, Severin SE. [Molecular mechanisms of niclosamide antitumor activity]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2016; 61:680-93. [PMID: 26716739 DOI: 10.18097/pbmc20156106680] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In this review the recent data regarding the antitumor activity of niclosamide and the molecular mechanisms of its antitumor activity are presented. Niclosamide has been used in the clinic for the treatment of intestinal parasite infections. In recent years in several screening investigations of various drugs and chemical compounds niclosamide was identified as a potential anticancer agent. Niclosamide not only inhibits the Wnt/β-catenin, mTORC1, STAT3, NF-κB and Notch signaling pathways, but also targets mitochondria in cancer cells to induce growth inhibition and apoptosis. A number of studies have established the anticancer activity of niclosamide in both in vitro and in vivo in xenotransplantation models using human tumors and immunodeficient mice. It is important that niclosamide is active not only against tumor cells but also cancer stem cells. Normal cells are resistant to niclosamide. The accumulated experimental data suggest niclosamide is a promising drug for the treatment of various types of cancer.
Collapse
Affiliation(s)
- E Yu Moskaleva
- National Research Centre "Kurchatov Institute", NBICS-Centre, Moscow, Russia
| | - V G Perevozchikova
- National Research Centre "Kurchatov Institute", NBICS-Centre, Moscow, Russia
| | - A S Zhirnik
- National Research Centre "Kurchatov Institute", NBICS-Centre, Moscow, Russia
| | - S E Severin
- National Research Centre "Kurchatov Institute", NBICS-Centre, Moscow, Russia
| |
Collapse
|
50
|
Repositioning of drugs for intervention in tumor progression and metastasis: Old drugs for new targets. Drug Resist Updat 2016; 26:10-27. [PMID: 27180307 DOI: 10.1016/j.drup.2016.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023]
Abstract
The increasing unraveling of the molecular basis of cancer offers manifold novel options for intervention strategies. However, the discovery and development of new drugs for potential clinical applications is a tremendously time-consuming and costly process. Translating a novel lead candidate compound into an approved clinical drug takes often more than a decade, and the success rate is very low due to versatile efforts including defining its pharmacokinetics, pharmacodynamics, side effects as well as lack of sufficient efficacy. Thus, strategies are needed to minimize time and costs, while maximizing success rates. A very attractive strategy for novel cancer therapeutic options is the repositioning of already approved drugs. These medicines, approved for the treatment of non-malignant disorders, have already passed some early costs and time, have been tested in humans and are ready for clinical trials as anti-cancer drugs. Here we discuss the repositioning of nonsteroidal anti-inflammatory drugs (NSAID), statins, anti-psychotic drugs, anti-helminthic drugs and vitamin D as anti-tumor agents. We focus on their novel actions and potential for inhibition of cancer growth and metastasis by interfering with target molecules and pathways, which drive these malignant processes. Furthermore, important pre-clinical and clinical data are reviewed herein, which elucidate their therapeutic mechanisms which enable their repositioning for cancer therapy and disruption of metastasis.
Collapse
|