601
|
NANDITHA CK, RAJAN MP. AN ADAPTIVE PHARMACOKINETIC OPTIMAL CONTROL APPROACH IN CHEMOTHERAPY FOR HETEROGENEOUS TUMOR. J BIOL SYST 2022. [DOI: 10.1142/s0218339022500188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The mathematical study of cancer received great attention in recent years. The theoretical and numerical investigations of models employed for studying the growth of cancer cells and treatment of cancer can help the clinical practitioners in adapting new strategies to face the challenges posed by the deadly disease. Chemotherapy is the most common method of treatment for cancer. The resistance of tumor cells toward the administered drug and the toxic effect of anti-cancer agents on healthy cells are major hurdles to the success of therapy. In this paper, we study this as an optimal control problem in which the amount of drug injected is taken as control. The evolution of different types of cells — sensitive tumor cells, resistant tumor cells and normal cells — under treatment are analyzed. The pharmacokinetics of the drug is also incorporated into the mathematical model. We propose a treatment protocol that assures the death of a maximum number of tumor cells but also manages to keep the normal cells at a sufficient level. We also validated our theoretical results numerically.
Collapse
Affiliation(s)
- C. K. NANDITHA
- School of Mathematics, Indian Institute of Science Education and Research Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, India
| | - M. P. RAJAN
- School of Mathematics, Indian Institute of Science Education and Research Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, India
| |
Collapse
|
602
|
Salihi A, Al-Naqshabandi MA, Khudhur ZO, Housein Z, Hama HA, Abdullah RM, Hussen BM, Alkasalias T. Gasotransmitters in the tumor microenvironment: Impacts on cancer chemotherapy (Review). Mol Med Rep 2022; 26:233. [PMID: 35616143 PMCID: PMC9178674 DOI: 10.3892/mmr.2022.12749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nitric oxide, carbon monoxide and hydrogen sulfide are three endogenous gasotransmitters that serve a role in regulating normal and pathological cellular activities. They can stimulate or inhibit cancer cell proliferation and invasion, as well as interfere with cancer cell responses to drug treatments. Understanding the molecular pathways governing the interactions between these gases and the tumor microenvironment can be utilized for the identification of a novel technique to disrupt cancer cell interactions and may contribute to the conception of effective and safe cancer therapy strategies. The present review discusses the effects of these gases in modulating the action of chemotherapies, as well as prospective pharmacological and therapeutic interfering approaches. A deeper knowledge of the mechanisms that underpin the cellular and pharmacological effects, as well as interactions, of each of the three gases could pave the way for therapeutic treatments and translational research.
Collapse
Affiliation(s)
- Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region 44001, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region 44002, Iraq
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Mohammed A. Al-Naqshabandi
- Department of Clinical Biochemistry, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region 44001, Iraq
| | - Zjwan Housein
- Department of Medical Laboratory Technology, Technical Health and Medical College, Erbil Polytechnique University, Erbil, Kurdistan Region 44002, Iraq
| | - Harmand A. Hama
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region 44002, Iraq
| | - Ramyar M. Abdullah
- College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Twana Alkasalias
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region 44002, Iraq
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
603
|
Annese T, Tamma R, Ribatti D. Update in TIGIT Immune-Checkpoint Role in Cancer. Front Oncol 2022; 12:871085. [PMID: 35656508 PMCID: PMC9152184 DOI: 10.3389/fonc.2022.871085] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
The in-depth characterization of cross-talk between tumor cells and T cells in solid and hematological malignancies will have to be considered to develop new therapeutical strategies concerning the reactivation and maintenance of patient-specific antitumor responses within the patient tumor microenvironment. Activation of immune cells depends on a delicate balance between activating and inhibitory signals mediated by different receptors. T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is an inhibitory receptor expressed by regulatory T cells (Tregs), activated T cells, and natural killer (NK) cells. TIGIT pathway regulates T cell-mediated tumor recognition in vivo and in vitro and represents an exciting target for checkpoint blockade immunotherapy. TIGIT blockade as monotherapy or in combination with other inhibitor receptors or drugs is emerging in clinical trials in patients with cancer. The purpose of this review is to update the role of TIGIT in cancer progression, looking at TIGIT pathways that are often upregulated in immune cells and at possible therapeutic strategies to avoid tumor aggressiveness, drug resistance, and treatment side effects. However, in the first part, we overviewed the role of immune checkpoints in immunoediting, the TIGIT structure and ligands, and summarized the key immune cells that express TIGIT.
Collapse
Affiliation(s)
- Tiziana Annese
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy.,Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
604
|
Alsaad H, Kubba A, Tahtamouni LH, Hamzah AH. Synthesis, docking study, and structure activity relationship of novel anti-tumor 1, 2, 4 triazole derivatives incorporating 2-(2, 3- dimethyl aminobenzoic acid) moiety. PHARMACIA 2022. [DOI: 10.3897/pharmacia.69.e83158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
A series of 1,2,4 triazole derivatives (H7-12) have been synthesized by reacting an excess of hydrazine hydrate with carbothioamide derivatives (H1-6). The final compounds (HB1-HB6) were synthesized by reacting the triazole derivatives with mefenamic acid using DCC as a coupling agent. The chemical structures were confirmed by FT-IR, 1H, and 13C-NMR spectra, and some physicochemical properties were determined. The cytotoxicity of the different compounds (HB1-HB6) was evaluated by the MTT assay against two human epithelial cancer cell lines, A549 lung carcinoma and Hep G2 hepatocyte carcinoma, and one normal human cell line WI-38 lung fibroblasts. The mode of cell killing (apoptosis versus necrosis), as well as the effect on cell cycle phases were evaluated via flow cytometry. Additionally, EGFR tyrosine kinase inhibition assay was performed. The results presented in the current study indicate that the six tested compounds exhibited cytotoxicity against both cancer cell lines, and the lowest IC50 was achieved with compound HB5 against Hep G2 cancer cells which was found to be highly selective against cancer cells. HB5-treated Hep G2 cells were arrested at the S and G2/M cell cycle phases. Compound HB5 caused cell killing via apoptosis rather than necrosis, and this was achieved by inhibiting EGFR tyrosine kinase activity needed for cell proliferation, and cell cycle progression. In silico pre-ADMET studies confirmed all final compounds don’t cause CNS side effects, with little liver dysfunction effect.
Collapse
|
605
|
Shi Z, Zheng J, Tang W, Bai Y, Zhang L, Xuan Z, Sun H, Shao C. Multifunctional Nanomaterials for Ferroptotic Cancer Therapy. Front Chem 2022; 10:868630. [PMID: 35402376 PMCID: PMC8987283 DOI: 10.3389/fchem.2022.868630] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/02/2022] [Indexed: 01/03/2023] Open
Abstract
Patient outcomes from the current clinical cancer therapy remain still far from satisfactory. However, in recent years, several biomedical discoveries and nanotechnological innovations have been made, so there is an impetus to combine these with conventional treatments to improve patient experience and disease prognosis. Ferroptosis, a term first coined in 2012, is an iron-dependent regulated cell death (RCD) based on the production of reactive oxygen species (ROS) and the consequent oxidization of polyunsaturated fatty acids (PUFAs). Many nanomaterials that can induce ferroptosis have been explored for applications in cancer therapy. In this review, we summarize the recent developments in ferroptosis-based nanomaterials for cancer therapy and discuss the future of ferroptosis, nanomedicine, and cancer therapy.
Collapse
Affiliation(s)
- Zhiyuan Shi
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jianzhong Zheng
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenbin Tang
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Bai
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lei Zhang
- School of Public Health, Xiamen Univerisity, Xiamen, China
| | - Zuodong Xuan
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Huimin Sun
- Central Laboratory, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Huimin Sun, ; Chen Shao,
| | - Chen Shao
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Huimin Sun, ; Chen Shao,
| |
Collapse
|
606
|
Mirian M, Hariri A, Yadollahi M, Kohandel M. Circadian and Immunity Cycle Talk in Cancer Destination: From Biological Aspects to In Silico Analysis. Cancers (Basel) 2022; 14:1578. [PMID: 35326729 PMCID: PMC8945968 DOI: 10.3390/cancers14061578] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is the leading cause of death and a major problem to increasing life expectancy worldwide. In recent years, various approaches such as surgery, chemotherapy, radiation, targeted therapies, and the newest pillar, immunotherapy, have been developed to treat cancer. Among key factors impacting the effectiveness of treatment, the administration of drugs based on the circadian rhythm in a person and within individuals can significantly elevate drug efficacy, reduce adverse effects, and prevent drug resistance. Circadian clocks also affect various physiological processes such as the sleep cycle, body temperature cycle, digestive and cardiovascular processes, and endocrine and immune systems. In recent years, to achieve precision patterns for drug administration using computational methods, the interaction of the effects of drugs and their cellular pathways has been considered more seriously. Integrated data-derived pathological images and genomics, transcriptomics, and proteomics analyses have provided an understanding of the molecular basis of cancer and dramatically revealed interactions between circadian and immunity cycles. Here, we describe crosstalk between the circadian cycle signaling pathway and immunity cycle in cancer and discuss how tumor microenvironment affects the influence on treatment process based on individuals' genetic differences. Moreover, we highlight recent advances in computational modeling that pave the way for personalized immune chronotherapy.
Collapse
Affiliation(s)
- Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran; (M.M.); (A.H.)
| | - Amirali Hariri
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran; (M.M.); (A.H.)
| | - Mahtasadat Yadollahi
- School of Pharmacology and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran;
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
607
|
Nur S, Aisyah AN, Lukitaningsih E, Rumiyati R, Fadri A, Marwati M. Cytotoxic Effect of the Paku Atai Merah (Angiopteris ferox Copel) Fraction on MCF-7 and HeLa Cells and its Compound Profile by GC-MS. BORNEO JOURNAL OF PHARMACY 2022. [DOI: 10.33084/bjop.v5i1.2943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cancer is a condition of abnormal cell proliferation of tissue cells in the body that becomes malignant. It can attack other parts of the body and affect the normal function of the body organs. The sample used in this study was tubers of paku atai merah (Angiopteris ferox Copel), then extracted using 96% ethanol eluent to obtain a thick extract. The ethanolic extract of A. ferox was fractionated using column chromatography to get the active fraction to characterize the compound using thin-layer chromatography and gas chromatography-mass spectroscopy (GC-MS) and tested its cytotoxic effectiveness on MCF-7 and HeLa cancer cells. The results of this study were obtained from fractionation using the column chromatography method to get sub-fraction C and the results of compound characterization using GC-MS and obtained variations in the class of compounds contained in the sample: amino acids, glucosinolates, alkaloids, flavonoids, and terpenoids. Based on the cytotoxic effect test of sub-fraction C on MCF-7 cells, the results obtained moderate cytotoxic effects with an IC50 value of 61.027 µg/mL, and HeLa cells had an IC50 value of 521.03 µg/mL, which was categorized as having a weak cytotoxic effect. Based on the results obtained from this study, it can be concluded that sub-fraction C of A. ferox tubers has a cytotoxic effect on MCF-7 cells to be used as a reference for tracing pure compounds from A. ferox tuber.
Collapse
|
608
|
Li K, Zhang Z, Mei Y, Li M, Yang Q, WU Q, Yang H, HE LIANGCAN, Liu S. Targeting innate immune system by nanoparticles for cancer immunotherapy. J Mater Chem B 2022; 10:1709-1733. [DOI: 10.1039/d1tb02818a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Various cancer therapies have advanced remarkably over the past decade. Unlike the direct therapeutic targeting of tumor cells, cancer immunotherapy is a new strategy that boosts the host's immune system...
Collapse
|