1
|
Rawat S, Moglad E, Afzal M, Goyal A, Roopashree R, Bansal P, Mishra S, Prasad GVS, Pramanik A, Alzarea SI, Ali H, Imran M, Abida. Reprogramming tumor-associated macrophages: The role of MEK-STAT3 inhibition in lung cancer. Pathol Res Pract 2025; 265:155748. [PMID: 39616977 DOI: 10.1016/j.prp.2024.155748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/10/2024] [Accepted: 11/27/2024] [Indexed: 12/11/2024]
Abstract
Tumor-associated macrophages (TAMs) crucially contribute to lung cancer's advancement and escape from the immune system. TAMs, particularly the M2 phenotype, promote an immunosuppressive microenvironment, facilitating tumor growth and metastasis. The MEK-STAT3 signalling pathway is a critical mediator in this process, driving TAM reprogramming and contributing to lung cancer's resistance to treatment. Inhibiting the MEK and STAT3 pathways disrupts key cancer-promoting mechanisms, including immune evasion, angiogenesis, and metastasis. Preclinical studies have demonstrated the effectiveness of MEK inhibitors, such as trametinib and selumetinib, in synergistic therapies for NSCLC, particularly in modulating the tumor microenvironment. We analyse the present understanding of approaches that can transform TAMs via the inhibition of MEK-STAT3 with either solo or combined treatments in lung cancer therapy.
Collapse
Affiliation(s)
- Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, PO Box 6231, Jeddah 21442, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - R Roopashree
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Shivang Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
2
|
Boța M, Vlaia L, Jîjie AR, Marcovici I, Crişan F, Oancea C, Dehelean CA, Mateescu T, Moacă EA. Exploring Synergistic Interactions between Natural Compounds and Conventional Chemotherapeutic Drugs in Preclinical Models of Lung Cancer. Pharmaceuticals (Basel) 2024; 17:598. [PMID: 38794168 PMCID: PMC11123751 DOI: 10.3390/ph17050598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
In the current work, the synergy between natural compounds and conventional chemotherapeutic drugs is comprehensively reviewed in light of current preclinical research findings. The prognosis for lung cancer patients is poor, with a 5-year survival rate of 18.1%. The use of natural compounds in combination with conventional chemotherapeutic drugs has gained significant attention as a potential novel approach in the treatment of lung cancer. The present work highlights the importance of finding more effective therapies to increase survival rates. Chemotherapy is a primary treatment option for lung cancer but it has limitations such as reduced effectiveness because cancer cells become resistant. Natural compounds isolated from medicinal plants have shown promising anticancer or chemopreventive properties and their synergistic effect has been observed when combined with conventional therapies. The combined use of an anti-cancer drug and a natural compound exhibits synergistic effects, enhancing overall therapeutic actions against cancer cells. In conclusion, this work provides an overview of the latest preclinical research on medicinal plants and plant-derived compounds as alternative or complementary treatment options for lung cancer chemotherapy and discusses the potential of natural compounds in treating lung cancer with minimal side effects.
Collapse
Affiliation(s)
- Mihaela Boța
- Department II—Pharmaceutical Technology, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (M.B.); (L.V.)
| | - Lavinia Vlaia
- Department II—Pharmaceutical Technology, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (M.B.); (L.V.)
- Formulation and Technology of Drugs Research Center, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Alex-Robert Jîjie
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Iasmina Marcovici
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Flavia Crişan
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Cristian Oancea
- Discipline of Pneumology, Department of Infectious Diseases, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania;
| | - Cristina Adriana Dehelean
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| | - Tudor Mateescu
- Department of Thoracic Surgery, Clinical Hospital for Infectious Diseases and Pneumophthiology Dr. Victor Babes, 13 Gheorghe Adam Street, RO-300310 Timisoara, Romania;
| | - Elena-Alina Moacă
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania; (I.M.); (F.C.); (C.A.D.); (E.-A.M.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, RO-300041 Timisoara, Romania
| |
Collapse
|
3
|
Wang Z, Li M, Bi L, Hu X, Wang Y. Traditional Chinese Medicine in Regulating Tumor Microenvironment. Onco Targets Ther 2024; 17:313-325. [PMID: 38617090 PMCID: PMC11016250 DOI: 10.2147/ott.s444214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024] Open
Abstract
Tumor microenvironment (TME) is a complex and integrated system containing a variety of tumor-infiltrating immune cells and stromal cells. They are closely connected with cancer cells and influence the development and progression of cancer. Traditional Chinese medicine (TCM) is an important complementary therapy for cancer treatment in China. It mainly eliminates cancer cells by regulating TME. The aim of this review is to systematically summarize the crosstalk between tumor cells and TME, and to summarize the research progress of TCM in regulating TME. The review is of great significance in revealing the therapeutic mechanism of action of TCM, and provides an opportunity for the combined application of TCM and immunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Ziwei Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Mengyao Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Ling Bi
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Xueqing Hu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
4
|
Jung T, Cheon C. Synergistic and Additive Effects of Herbal Medicines in Combination with Chemotherapeutics: A Scoping Review. Integr Cancer Ther 2024; 23:15347354241259416. [PMID: 38867515 PMCID: PMC11179546 DOI: 10.1177/15347354241259416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Natural products are increasingly gaining interest as potential new drug candidates for cancer treatment. Herbal formula, which are combinations of several herbs, are primarily used in East Asia and have a long history of use that continues today. Recently, research exploring the combination of herbal formulas and chemotherapy for cancer treatment has been on the rise. METHODS This study reviewed research on the co-administration of herbal formulas and chemotherapy for cancer treatment. The databases PubMed, Embase, and Cochrane Library were used for article searches. The following keywords were employed: "Antineoplastic agents," "Chemotherapy," "Phytotherapy," "Herbal medicine," "Drug synergism," and "Synergistic effect." The selection process focused on studies that investigated the synergistic interaction between herbal formulas and chemotherapeutic agents. RESULTS Among the 30 studies included, 25 herbal formulas and 7 chemotherapies were used. The chemotherapy agents co-administered included cisplatin, 5-fluorouracil, docetaxel, doxorubicin, oxaliplatin, irinotecan, and gemcitabine. The types of cancer most frequently studied were lung, breast, and colon cancers. Most studies evaluating the anticancer efficacy of combined herbal formula and chemotherapy treatment were conducted in vitro or in vivo. DISCUSSION Most studies reported synergistic effects on cytotoxicity, apoptosis, and tumor growth inhibition. These effects were found to be associated with cell cycle arrest, anti-angiogenesis, and gene expression regulation. Further studies leading to clinical trials are required. Clinical experiences in East Asian countries could provide insights for future research.
Collapse
Affiliation(s)
- Taehun Jung
- Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Chunhoo Cheon
- Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| |
Collapse
|
5
|
Mendez-Callejas G, Piñeros-Avila M, Yosa-Reyes J, Pestana-Nobles R, Torrenegra R, Camargo-Ubate MF, Bello-Castro AE, Celis CA. A Novel Tri-Hydroxy-Methylated Chalcone Isolated from Chromolaena tacotana with Anti-Cancer Potential Targeting Pro-Survival Proteins. Int J Mol Sci 2023; 24:15185. [PMID: 37894866 PMCID: PMC10607159 DOI: 10.3390/ijms242015185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Chromolaena tacotana (Klatt) R. M. King and H. Rob (Ch. tacotana) contains bioactive flavonoids that may have antioxidant and/or anti-cancer properties. This study investigated the potential anti-cancer properties of a newly identified chalcone isolated from the inflorescences of the plant Chromolaena tacotana (Klatt) R. M. King and H. Rob (Ch. tacotana). The chalcone structure was determined using HPLC/MS (QTOF), UV, and NMR spectroscopy. The compound cytotoxicity and selectivity were evaluated on prostate, cervical, and breast cancer cell lines using the MTT assay. Apoptosis and autophagy induction were assessed through flow cytometry by detecting annexin V/7-AAD, active Casp3/7, and LC3B proteins. These results were supported by Western blot analysis. Mitochondrial effects on membrane potential, as well as levels of pro- and anti-apoptotic proteins were analyzed using flow cytometry, fluorescent microscopy, and Western blot analysis specifically on a triple-negative breast cancer (TNBC) cell line. Furthermore, molecular docking (MD) and molecular dynamics (MD) simulations were performed to evaluate the interaction between the compounds and pro-survival proteins. The compound identified as 2',3,4-trihydroxy-4',6'-dimethoxy chalcone inhibited the cancer cell line proliferation and induced apoptosis and autophagy. MDA-MB-231, a TNBC cell line, exhibited the highest sensitivity to the compound with good selectivity. This activity was associated with the regulation of mitochondrial membrane potential, activation of the pro-apoptotic proteins, and reduction of anti-apoptotic proteins, thereby triggering the intrinsic apoptotic pathway. The chalcone consistently interacted with anti-apoptotic proteins, particularly the Bcl-2 protein, throughout the simulation period. However, there was a noticeable conformational shift observed with the negative autophagy regulator mTOR protein. Future studies should focus on the molecular mechanisms underlying the anti-cancer potential of the new chalcone and other flavonoids from Ch. tacotana, particularly against predominant cancer cell types.
Collapse
Affiliation(s)
- Gina Mendez-Callejas
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada (GIBGA), Laboratorio de Biología Celular y Molecular, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia;
| | - Marco Piñeros-Avila
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada (GIBGA), Laboratorio de Biología Celular y Molecular, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia;
| | - Juvenal Yosa-Reyes
- Grupo de Investigación en Ciencias Exactas, Física y Naturales Aplicadas, Facultad de Ciencias Básicas y Biomédicas, Laboratorio de Simulación Molecular y Bioinformática, Universidad Simón Bolívar, Carrera 59 # 59-65, Barranquilla 080002, Colombia; (J.Y.-R.)
| | - Roberto Pestana-Nobles
- Grupo de Investigación en Ciencias Exactas, Física y Naturales Aplicadas, Facultad de Ciencias Básicas y Biomédicas, Laboratorio de Simulación Molecular y Bioinformática, Universidad Simón Bolívar, Carrera 59 # 59-65, Barranquilla 080002, Colombia; (J.Y.-R.)
| | - Ruben Torrenegra
- Grupo de Investigación en Productos Naturales de la U.D.C.A. (PRONAUDCA), Laboratorio de Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia
| | - María F. Camargo-Ubate
- Grupo de Investigación en Productos Naturales de la U.D.C.A. (PRONAUDCA), Laboratorio de Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia
| | - Andrea E. Bello-Castro
- Grupo de Investigación en Productos Naturales de la U.D.C.A. (PRONAUDCA), Laboratorio de Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia
| | - Crispin A. Celis
- Grupo de Investigación en Fitoquímica (GIFUJ), Departamento de Química, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 # 40-62, Bogotá 1115511, Colombia
| |
Collapse
|
6
|
Tan N, Zhao W, Wang Y, Li P, Liu J, Sun Z, Pan J, Song S, Li S, Liu Z, Bian Y. AHR, a novel inhibitory immune checkpoint receptor, is a potential therapeutic target for chemoresistant glioblastoma. J Cancer Res Clin Oncol 2023; 149:9705-9720. [PMID: 37233762 DOI: 10.1007/s00432-023-04894-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/20/2023] [Indexed: 05/27/2023]
Abstract
PURPOSE This study aims to elucidate the mechanism underlying temozolomide resistance in patients with MGMT promoter hypomethylated glioblastoma, which is correlated with poor prognosis. The objective is to identify therapeutic targets and drugs suitable for temozolomide-resistant glioblastoma patients using big data analysis. METHODS In this retrospective study, transcriptome sequencing data from 457 glioblastoma patients, multi-omics data, and single-cell sequencing data were employed to assess the expression pattern, prognostic value, and biological functions of AHR in glioblastoma. The HERB database was utilized to screen for AHR-targeted drugs for glioblastoma treatment. Validation of our findings was conducted using multiplex immunofluorescence staining of clinical samples and T cells and tumor cells co-culture models. RESULTS Our findings demonstrated that patients with MGMT promoter unmethylation did not benefit from postoperative temozolomide chemotherapy due to resistance arising from DNA repair function and tumor immune response. AHR was found to be expressed in immune cells and exhibited an immunomodulatory role in glioblastoma with MGMT promoter unmethylation. AHR was identified as a potential novel inhibitory immune checkpoint receptor, serving as a therapeutic target for temozolomide-resistant glioblastoma. Furthermore, targeting AHR with Semen aesculi markedly enhanced the cytotoxic effect of T cells on glioma cells. CONCLUSIONS In addition to DNA repair function, the tumor immune response plays a pivotal role in temozolomide resistance of glioblastoma. Herbal compounds targeting AHR may offer an effective treatment for temozolomide-resistant glioblastoma.
Collapse
Affiliation(s)
- Nian Tan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China.
| | - Wei Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Yiyang Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Ping Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Jianwei Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Zhaoying Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Jianming Pan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Shilin Song
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Shunyao Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Ziyi Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, People's Republic of China.
| |
Collapse
|
7
|
Tang P, Shen T, Wang H, Zhang R, Zhang X, Li X, Xiao W. Challenges and opportunities for improving the druggability of natural product: Why need drug delivery system? Biomed Pharmacother 2023; 164:114955. [PMID: 37269810 DOI: 10.1016/j.biopha.2023.114955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/14/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023] Open
Abstract
Bioactive natural products (BNPs) are the marrow of medicinal plants, which are the secondary metabolites of organisms and have been the most famous drug discovery database. Bioactive natural products are famous for their enormous number and great safety in medical applications. However, BNPs are troubled by their poor druggability compared with synthesis drugs and are challenged as medicine (only a few BNPs are applied in clinical settings). In order to find a reasonable solution to improving the druggability of BNPs, this review summarizes their bioactive nature based on the enormous pharmacological research and tries to explain the reasons for the poor druggability of BNPs. And then focused on the boosting research on BNPs loaded drug delivery systems, this review further concludes the advantages of drug delivery systems on the druggability improvement of BNPs from the perspective of their bioactive nature, discusses why BNPs need drug delivery systems, and predicts the next direction.
Collapse
Affiliation(s)
- Peng Tang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, China; School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China; Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
| | - Tianze Shen
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, China; School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China; Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
| | - Hairong Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, China; School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China; Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
| | - Ruihan Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, China; School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China; Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
| | - Xingjie Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, China; School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China; Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
| | - Xiaoli Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, China; School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China; Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China.
| | - Weilie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan University, Kunming, China; School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China; Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China.
| |
Collapse
|
8
|
Xie C, Liang C, Wang R, Yi K, Zhou X, Li X, Chen Y, Miao D, Zhong C, Zhu J. Resveratrol suppresses lung cancer by targeting cancer stem-like cells and regulating tumor microenvironment. J Nutr Biochem 2023; 112:109211. [PMID: 36370924 DOI: 10.1016/j.jnutbio.2022.109211] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 06/30/2022] [Accepted: 09/23/2022] [Indexed: 11/11/2022]
Abstract
Increasing evidence indicate that cancer stem cells (CSCs) are the key driver of tumor initiation and recurrence. The cellular and soluble components of the tumor microenvironment (TME) impact on cancer initiation and progression, such as cytokines and chemokines. Thus, targeting CSCs and TME is a novel anti-cancer approach. Resveratrol (RES), a bioactive phytochemical extracted from various plants, exhibits tumor-suppressing activities in lung cancer, yet the mechanism remains poorly understood. Our data showed that the expression level of IL-6 was positively correlated with the presence of lung cancer stem-like cells (LCSCs) in human lung cancer tissues. In vitro results showed that IL-6 was highly elevated in lung cancer sphere-forming cells and could enhance the stemness of LCSCs, including tumor sphere formation ability, the percentage of CD133 positive cells, and the expression of LCSC specific markers (CD133, ALDH1A1 and Nanog). Simultaneously, our results confirmed that RES effectively inhibited LCSC properties, downregulated Wnt/β-catenin signaling and reduced IL-6 level in vitro and in vivo. Furthermore, we found RES treatment attenuated the activation of Wnt/β-catenin signaling by LiCl (GSK3β agonist). IL-6-promoted LCSC properties and Wnt/β-catenin signaling was also reversed by RES. Taken together, these data illustrated that RES inhibited lung cancer by targeting LCSCs and IL-6 in TME. The novel findings from this study provided evidence that RES exhibited multi-target effects on suppression of lung cancer and could be a novel potent cancer-preventive compound.
Collapse
Affiliation(s)
- Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunhua Liang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rong Wang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kefan Yi
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Zhou
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoting Li
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Chen
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dengshun Miao
- Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Cancer Research Division, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jianyun Zhu
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| |
Collapse
|
9
|
Reveal the Mechanisms of Yi-Fei-Jian-Pi-Tang on Covid-19 through Network Pharmacology Approach. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:1493137. [PMID: 35855804 PMCID: PMC9288182 DOI: 10.1155/2022/1493137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/01/2022] [Indexed: 11/19/2022]
Abstract
Objectives The Traditional Chinese Medicine (TCM) formula Yi-Fei-Jian-Pi-Tang (YFJPT) has been demonstrated effective against Corona Virus Disease 2019 (Covid-19). The aim of this article is to make a thorough inquiry about its active constituent as well as mechanisms against Covid-19 via TCM network pharmacology. Methods All the ingredients of YFJPT are obtained from the pharmacology database of the TCM system. The genes which are associated with the targets are obtained by utilizing UniProt. The herb-target network is built up by utilizing Cytoscape. The target protein-protein interaction network is built by utilizing the STRING database and Cytoscape. The critical targets of YFJPT are explored by Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Results The outcomes show that YFJPT might has 33 therapeutic targets on Covid-19, namely, interleukin 2 (IL2), heme oxygenase 1 (HMOX1), interleukin 4 (IL4), interferon gamma (FNG), α nuclear factor of kappa light polypeptide gene enhancer in Bcells inhibitor, alpha (NFKBIA), nuclear factor-k-gene binding (NFKB), nitric oxide synthase 3 (NOS3), intercellular adhesion molecule 1 (ICAM1), hypoxia inducible factor 1 subunit alpha (HIF1A), mitogen-activated protein kinase 3 (MAPK3), epidermal growth factor receptor (EGFR), interleukin 10 (IL10), jun proto-oncogene (JUN), C-C motif chemokine ligand 2 (CCL2), C-X-C motif chemokine ligand 8 (CXCL8), tumor protein p53 (TP53), interleukin 1 beta (IL1B), AKT serine/threonine kinase 1 (AKT1), tumor necrosis factor (TNF), interleukin 6 (IL6), erb-b2 receptor tyrosine kinase 2 (ERBB2), RELA proto-oncogene (RELA), NF-κB subunit, caspase 8 (CASP8), peroxisome proliferator activated receptor alpha (PPARA), TIMP metallopeptidase inhibitor 1 (TIMP1), transforming growth factor beta 1 (TGFB1), interleukin 1 alpha (IL1A), signal transducer and activator of transcription 1 (STAT1), mitogen-activated protein kinase 8 (MAPK8), myeloperoxidase (MPO), matrix metallopeptidase 3 (MMP3), matrix metallopeptidase 1 (MMP1), and NFE2 like bZIP transcription factor 2 (NFE2L2). The gene enrichment analysis prompts that YFJPT most likely contributes to patients related to Covid-19 by regulating the pathways of cancers. Conclusions That will lay a foundation for the clinical rational application and further experimental research of YFJPT.
Collapse
|
10
|
Modeling the efficacy of different anti-angiogenic drugs on treatment of solid tumors using 3D computational modeling and machine learning. Comput Biol Med 2022; 146:105511. [DOI: 10.1016/j.compbiomed.2022.105511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/11/2022]
|
11
|
Dini I. The commercial importance to develop validated analytical methods to define phytochemical levels in herbal medicinal products. Phytother Res 2022; 36:3675-3677. [PMID: 35537696 DOI: 10.1002/ptr.7485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Irene Dini
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Chen S, Nishi M, Morine Y, Shimada M, Tokunaga T, Kashihara H, Takasu C, Yamada S, Wada Y. Epigallocatechin‑3‑gallate hinders metabolic coupling to suppress colorectal cancer malignancy through targeting aerobic glycolysis in cancer‑associated fibroblasts. Int J Oncol 2022; 60:19. [PMID: 35029285 PMCID: PMC8776327 DOI: 10.3892/ijo.2022.5309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
In recent times, researchers working on tumor metabolism have paid increasing attention to the tumor microenvironment. Emerging evidence has confirmed that epigenetic modifications of cancer-associated fibroblasts (CAFs) alters the characteristics of glucose metabolism to achieve a symbiotic relationship with the cancer cells. Epigallocatechin-3-gallate (EGCG) exerts anti-tumor effects via a variety of mechanisms, although the underlying mechanism that accounts for the effects of EGCG on glucose metabolic alterations of CAFs have yet to be elucidated. In the present study, through co-culture with colorectal cancer (CRC) cells, human intestinal fibroblasts were transformed into CAFs, and exhibited enhanced aerobic glycolysis. Induced CAFs were able to enhance the proliferation, migration and invasion of CRC cells in vitro. EGCG treatment led to direct inhibition of the proliferation and migration of CRC cells; furthermore, EGCG treatment of CAFs suppressed their tumor-promoting capabilities by inhibiting their glycolytic activity. Blocking the lactic acid efflux of CAFs with a monocarboxylate transporter 4 (MCT4) inhibitor or through silencing MCT4 could also suppress their tumor-promoting capabilities, indicating that lactate fulfills an important role in the metabolic coupling that occurs between CAFs and cancer cells. Taken together, the results of the present study showed that EGCG targeting of the metabolism of tumor stromal cells provided a safe and effective strategy of anti-cancer therapy.
Collapse
Affiliation(s)
- Shuhai Chen
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Masaaki Nishi
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Yuji Morine
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Mitsuo Shimada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Takuya Tokunaga
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Hideya Kashihara
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Chie Takasu
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Shinichiro Yamada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| | - Yuma Wada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University, Tokushima 770‑8503, Japan
| |
Collapse
|
13
|
de Souza PS, Bibá GCC, Melo EDDN, Muzitano MF. Chalcones against the hallmarks of cancer: a mini-review. Nat Prod Res 2021; 36:4809-4826. [PMID: 34865580 DOI: 10.1080/14786419.2021.2000980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Chalcones (1,3-diphenylpropen-1-ones) are a class of flavonoids that have been shown a broad spectrum of biological activities with therapeutic potential. Naturally occurring chalcones or synthetic chalcone derivatives have been extensively investigated as anticancer compounds. Cancer is still among the leading causes of death globally, although cancer treatments have improved over the past decades. Most of chemotherapeutic drugs target proliferating tumor cells; however, the cancer cells capabilities are also associated to tumor surround microenvironment. Thereby, the search of new compounds with a broad antitumor activity is still a great challenge. The cytotoxicity mechanisms of chalcones are beyond apoptosis induction in tumor cells, which make them promising compound for cancer therapy. In this mini-review we summarized recent studies that describe the anticancer potential of chalcones related to some of hallmarks of cancer. We shed a light on sustaining proliferative signaling, tumor-promoting inflammation, activating invasion and metastasis, inducing angiogenesis and resisting cell death.
Collapse
Affiliation(s)
- Paloma Silva de Souza
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Geysa Cristina Caldas Bibá
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelynn Dalila do Nascimento Melo
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle Frazão Muzitano
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Cheon C. Synergistic effects of herbal medicines and anticancer drugs: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e27918. [PMID: 34797348 PMCID: PMC8601363 DOI: 10.1097/md.0000000000027918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND An increasing number of studies have been reporting combination therapy using herbal medicines and anticancer drugs, and the synergistic effects of this combination have gained much attention across the medical community. In this study, we will review and summarize all published studies that have investigated the synergistic interaction between herbal medicines and anticancer drugs. METHODS We will search the PubMed, Embase, and Cochrane Library databases. Studies investigated the synergistic interaction between herbal medicines and anticancer drugs will be included. The selection and extraction process will be performed by 2 independent reviewers, and we will perform qualitative synthesis. DISCUSSION The present study is being performed to investigate the herbal medicines and anticancer drugs that are used concomitantly, and to determine the combinations that are expected to show a synergistic effect. This knowledge will provide new insights into the possible role of herbal medicines in anticancer treatment. REVIEW REGISTRATION Trial registration: OSF Registration number: DOI 10.17605/OSF.IO/H5QS9.
Collapse
|
15
|
The curcumin analog EF24 inhibits proliferation and invasion of triple-negative breast cancer cells by targeting the lncRNA HCG11/Sp1 axis. Mol Cell Biol 2021; 42:e0016321. [PMID: 34780286 DOI: 10.1128/mcb.00163-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
EF24, a curcumin analog, exerts a potent anti-tumor effect on various cancers. However, whether EF24 retards the progression of triple-negative breast cancer (TNBC) remains unclear. In this study, we explored the role of EF24 in TNBC and clarified the underlying mechanism. In a mouse model of TNBC xenograft, EF24 administration reduced the tumor volume, suppressed cell proliferation, promoted cell apoptosis, and downregulated long non-coding RNA human leukocyte antigen complex group 11 (HCG11) expression. In TNBC cell lines, EF24 administration reduced cell viability, suppressed cell invasion, and downregulated HCG11 expression. HCG11 overexpression re-enhanced the proliferation and invasion of TNBC cell lines suppressed by EF24. The following mechanism research revealed that HCG11 overexpression elevated Sp1 transcription factor (Sp1) expression by reducing its ubiquitination, thereby enhanced Sp1-mediated cell survival and invasion in the TNBC cell line. Finally, the in vivo study showed that HCG11-overexpressed TNBC xenografts exhibited lower responsiveness in response to EF24 treatment. In conclusion, EF24 treatment reduced HCG11 expression, resulting in the degradation of Sp1 expression, thereby inhibiting the proliferation and invasion of TNBC cells.
Collapse
|
16
|
Circulating Tumour Cells (CTCs) in NSCLC: From Prognosis to Therapy Design. Pharmaceutics 2021; 13:pharmaceutics13111879. [PMID: 34834295 PMCID: PMC8619417 DOI: 10.3390/pharmaceutics13111879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023] Open
Abstract
Designing optimal (neo)adjuvant therapy is a crucial aspect of the treatment of non-small-cell lung carcinoma (NSCLC). Standard methods of chemotherapy, radiotherapy, and immunotherapy represent effective strategies for treatment. However, in some cases with high metastatic activity and high levels of circulating tumour cells (CTCs), the efficacy of standard treatment methods is insufficient and results in treatment failure and reduced patient survival. CTCs are seen not only as an isolated phenomenon but also a key inherent part of the formation of metastasis and a key factor in cancer death. This review discusses the impact of NSCLC therapy strategies based on a meta-analysis of clinical studies. In addition, possible therapeutic strategies for repression when standard methods fail, such as the administration of low-toxicity natural anticancer agents targeting these phenomena (curcumin and flavonoids), are also discussed. These strategies are presented in the context of key mechanisms of tumour biology with a strong influence on CTC spread and metastasis (mechanisms related to tumour-associated and -infiltrating cells, epithelial–mesenchymal transition, and migration of cancer cells).
Collapse
|
17
|
Malla RR, Padmaraju V, Marni R, Kamal MA. Natural products: Potential targets of TME related long non-coding RNAs in lung cancer. PHYTOMEDICINE 2021; 93:153782. [PMID: 34627097 DOI: 10.1016/j.phymed.2021.153782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/08/2021] [Accepted: 09/26/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Lung cancer is a significant health concern worldwide due to high mortality and morbidity, despite the advances in diagnosis, treatment, and management. Recent experimental evidence from different models suggested long non-coding RNAs (lncRNAs) as major modulators of cancer stem cells (CSCs) in the tumor microenvironment (TME) to support metastasis and drug resistance in lung cancer. Evidence-based studies demonstrated that natural products interfere with TME functions. PURPOSE OF STUDY To establish lncRNAs of TME as novel targets of natural compounds for lung cancer management. STUDY DESIGN Current study used a combination of TME and lung CSCs, lncRNAs and enrichment and stemness maintenance, natural products and stem cell management, natural products and lncRNAs, natural products and targeted delivery as keywords to retrieve the literature from Scopus, Web of Science, PubMed, and Google Scholar. This study critically reviewed the current literature and presented cancer stem cells' ability in reprogramming lung TME. RESULTS This review found that TME related oncogenic and tumor suppressor lncRNAs and their signaling pathways control the maintenance of stemness in lung TME. This review explored natural phenolic compounds and found that curcumin, genistein, quercetin epigallocatechin gallate and ginsenoside Rh2 are efficient in managing lung CSCs. They modulate lncRNAs and their upstream mediators by targeting signaling and epigenetic pathways. This review also identified relevant nanotechnology-based phytochemical delivery approaches for targeting lung cancer. CONCLUSION By critical literature analysis, TME related lncRNAs were identified as potential therapeutic targets, aiming to develop natural product-based therapeutics to treat metastatic and drug-resistant lung cancers.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India; Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be) University, Visakhapatnam, Andhra Pradesh 530045, India.
| | - Vasudevaraju Padmaraju
- Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be) University, Visakhapatnam, Andhra Pradesh 530045, India
| | - Rakshmitha Marni
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India; Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be) University, Visakhapatnam, Andhra Pradesh 530045, India
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, Novel Global Community Educational Foundation, Australia
| |
Collapse
|
18
|
Wu Z, Zhang C, Najafi M. Targeting of the tumor immune microenvironment by metformin. J Cell Commun Signal 2021; 16:333-348. [PMID: 34611852 DOI: 10.1007/s12079-021-00648-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Stimulating antitumor immunity is an attractive idea for suppressing tumors. CD4 + and CD8 + T cells as well as natural killer cells (NK) are the primary antitumor immune cells in the tumor microenvironment (TME). In contrast to these cells, regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs) release several molecules to suppress antitumor immunity and stimulate cancer cell invasion and proliferation. Adjuvant treatment with certain nontoxic agents is interesting to boost antitumor immunity. Metformin, which is known as an antidiabetes drug, can modulate both antitumor and protumor immune cells within TME. It has the ability to induce the proliferation of CD8 + T lymphocytes and NK cells. On the other hand, metformin attenuates polarization toward TAMs, CAFs, and Tregs. Metformin also may stimulate the antitumor activity of immune system cells, while it interrupts the positive cross-talk and interactions between immunosuppressive cells and cancer cells. The purpose of this review is to explain the basic mechanisms for the interactions and communications between immunosuppressive, anti-tumoral, and cancer cells within TME. Next, we discuss the modulating effects of metformin on various cells and secretions in TME.
Collapse
Affiliation(s)
- Zihong Wu
- Department of Oncology, The NO.3 People's Hospital of Hubei Province, Jianghan University, Wuhan, 430033, Hubei, China
| | - Caidie Zhang
- Emergency Department, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, 430014, Hubei, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Chen L, Musa AE. Boosting immune system against cancer by resveratrol. Phytother Res 2021; 35:5514-5526. [PMID: 34101276 DOI: 10.1002/ptr.7189] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/03/2021] [Accepted: 05/26/2021] [Indexed: 01/16/2023]
Abstract
Modulation of the immune system is a critical part of anticancer therapies including immunotherapy, chemotherapy, and radiotherapy. The aim of immunomodulation in cancer therapy is boosting immune system cells including CD8+ T lymphocytes and natural killer (NK) cells, as well as suppression of immunosuppressive responses by macrophages and regulatory T cells (Tregs). Usually, using single or dual modality can induce immune system responses against cancer. However, immunosuppressive responses attenuate antitumor immunity following cancer therapy. Using some agents to boost immune system's function against cancer can increase therapeutic efficiency of anticancer therapy. Resveratrol, as a natural agent, has shown ability to modulate the immune system to potentiate antitumor immunity. Resveratrol has been shown to induce the release of anticancer cytokines such as IFN-γ and TNF-α and also inhibits the release of TGF-β. It also can stimulate the polarization of CD4+ T cells and macrophages toward anticancer cells and reduce infiltration and polarization of immunosuppressive cells. Furthermore, resveratrol can sensitize cancer cells to the released dead signals by anticancer immune cells. This review explains how resveratrol can boost the immune system against cancer via modulation of immune cell responses within tumor.
Collapse
Affiliation(s)
- Libo Chen
- School of Pharmaceutical and Environmental Technology, Jilin Vocational College of Industry and Technology, Jilin, China
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|