51
|
Valencia-Lazcano AA, Hassan D, Pourmadadi M, Shamsabadipour A, Behzadmehr R, Rahdar A, Medina DI, Díez-Pascual AM. 5-Fluorouracil nano-delivery systems as a cutting-edge for cancer therapy. Eur J Med Chem 2023; 246:114995. [PMID: 36493619 DOI: 10.1016/j.ejmech.2022.114995] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
5-Fluorouracil (5-FU) is amongst the most commonly used antimetabolite chemotherapeutic agents in recent decades. However, its low bioavailability, short half-life, rapid metabolism and the development of drug resistance after chemotherapy limit its therapeutic efficiency. In this study, 5-FU applications as an anti-cancer drug for treating diverse types of cancers (e.g. colon, pancreatic and breast) have been reviewed. Different approaches lately designed to circumvent the drawbacks of 5-FU therapy are described herein, including 5-FU-loaded lipid-based nanoparticles (NPs), polymeric NPs (both stimuli and non-stimuli responsive), carbon-based nanostructures and inorganic NPs. Furthermore, co-delivery systems of 5-FU with other drugs (e.g. paclitaxel, gelatin-doxorubicin and naproxen) have been reviewed, which aid to attain better bioavailability, higher effectiveness at a lower concentration and lower toxicity. This review provides researchers with the latest progress on 5-FU-loaded nanocarriers, which show great potential as an advanced tool for cancer therapy.
Collapse
Affiliation(s)
| | - Dilawar Hassan
- Tecnologico de Monterrey, School of Engineering and Sciences, Atizapan de Zaragoza, Estado de Mexico 52926, Mexico.
| | - Mehrab Pourmadadi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Amin Shamsabadipour
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Razieh Behzadmehr
- Department of Radiology, Zabol University of Medical Sciences, Zabol, Iran.
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, 538-98615, Zabol, Iran.
| | - Dora I Medina
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Nuevo Leon 64849, Monterrey, Mexico.
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805, Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
52
|
Sharma A, Shambhwani D, Pandey S, Singh J, Lalhlenmawia H, Kumarasamy M, Singh SK, Chellappan DK, Gupta G, Prasher P, Dua K, Kumar D. Advances in Lung Cancer Treatment Using Nanomedicines. ACS OMEGA 2023; 8:10-41. [PMID: 36643475 PMCID: PMC9835549 DOI: 10.1021/acsomega.2c04078] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/13/2022] [Indexed: 06/01/2023]
Abstract
Carcinoma of the lungs is among the most menacing forms of malignancy and has a poor prognosis, with a low overall survival rate due to delayed detection and ineffectiveness of conventional therapy. Therefore, drug delivery strategies that may overcome undesired damage to healthy cells, boost therapeutic efficacy, and act as imaging tools are currently gaining much attention. Advances in material science have resulted in unique nanoscale-based theranostic agents, which provide renewed hope for patients suffering from lung cancer. Nanotechnology has vastly modified and upgraded the existing techniques, focusing primarily on increasing bioavailability and stability of anti-cancer drugs. Nanocarrier-based imaging systems as theranostic tools in the treatment of lung carcinoma have proven to possess considerable benefits, such as early detection and targeted therapeutic delivery for effectively treating lung cancer. Several variants of nano-drug delivery agents have been successfully studied for therapeutic applications, such as liposomes, dendrimers, polymeric nanoparticles, nanoemulsions, carbon nanotubes, gold nanoparticles, magnetic nanoparticles, solid lipid nanoparticles, hydrogels, and micelles. In this Review, we present a comprehensive outline on the various types of overexpressed receptors in lung cancer, as well as the various targeting approaches of nanoparticles.
Collapse
Affiliation(s)
- Akshansh Sharma
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| | | | - Sadanand Pandey
- Department
of Chemistry, College of Natural Sciences, Yeungnam University, Gyeongsan, Gyeongbuk 38541, South Korea
| | - Jay Singh
- Department
of Chemistry, Institute of Science, Banaras
Hindu University, Varanasi 221005, India
| | - Hauzel Lalhlenmawia
- Department
of Pharmacy, Regional Institute of Paramedical
and Nursing Sciences, Zemabawk, Aizawl, Mizoram 796017, India
| | - Murali Kumarasamy
- Department
of Biotechnology, National Institute of
Pharmaceutical Education and Research, Hajipur 844102, India
| | - Sachin Kumar Singh
- School
of Pharmaceutical Sciences, Lovely Professional
University, Phagwara 144411, India
- Faculty
of Health, Australian Research Centre in Complementary and Integrative
Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department
of Life Sciences, School of Pharmacy, International
Medical University, Kuala Lumpur 57000, Malaysia
| | - Gaurav Gupta
- Department
of Pharmacology, School of Pharmacy, Suresh
Gyan Vihar University, Jaipur 302017, India
- Department
of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical
and Technical Sciences, Saveetha University, Chennai 602117, India
- Uttaranchal
Institute of Pharmaceutical Sciences, Uttaranchal
University, Dehradun 248007, India
| | - Parteek Prasher
- Department
of Chemistry, University of Petroleum &
Energy Studies, Dehradun 248007, India
| | - Kamal Dua
- Faculty
of Health, Australian Research Centre in Complementary and Integrative
Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- Discipline
of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Deepak Kumar
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| |
Collapse
|
53
|
Sharma V, Dhawan S, Kumar A, Kaur J. P19 a Parthenin Analog Induces Cell Lineage Dependent Apoptotic and Immunomodulatory Signaling in Acute Lymphoid Leukemia Cells. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2023; 12:1-17. [PMID: 37942260 PMCID: PMC10629723 DOI: 10.22088/ijmcm.bums.12.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/30/2023] [Accepted: 09/30/2023] [Indexed: 11/10/2023]
Abstract
Leukemia is a type of cancer that affects the blood and bone marrow. Acute lymphoid leukaemia, also known as ALL, is regarded as one of the deadliest forms of cancer. Due to the rapid increase in various cancer cases and the development of resistance in cancer cells, it is necessary to identify novel lead molecules with more potent anticancer properties. There is a growing interest in using herbal products/analogs as multi-component agents (as anticancer agents and immunomodulators) for cancer treatment. In the present investigation, an attempt has been made to explore the anticancer and immunomodulatory activity of P19, an analog of parthenin in ALL. P19 was reported to exhibit anticancer efficacy by triggering apoptotic signaling events in human leukaemia HL-60 cells by significant NO production. In contrast to this finding, ROS and NO were not required for P19-mediated apoptosis in Raji cells. The mechanism of action of P19 was observed to be cancer cell lineage dependent. P19 demonstrated very effective anticancer properties against ALL (IC50 3µM). Molecular investigations revealed that P19 induced mitochondrion mediated apoptosis by Bax localization to mitochondria and enhanced cytosolic calcium in the cytoplasm. Further activation of the caspase 3, caspase 8 and PARP cleavage suggested the involvement of the caspase-mediated apoptosis. Anti-proliferative activity revealed the telomerase inhibition and cell cycle arrest in G0/G1 phase after P19 treatment. Immunomodulatory effects of the P19 revealed the enhanced INFɣ and NO production in Jurkat and THP cells. Owing to its antiproliferative and immunomodulatory potential against leukemia cells P19 can further be explored as effective therapeutics against leukemia.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Biotechnology, Panjab University, Chandigarh, India.
| | - Samriti Dhawan
- Department of Biotechnology, Goswami Ganesh Dutta Sanatan Dharma College, Chandigarh, India.
| | - Ajay Kumar
- Pharmacology Division, Indian Institute of Integrative Medicine, Jammu, India.
| | - Jagdeep Kaur
- Department of Biotechnology, Panjab University, Chandigarh, India.
| |
Collapse
|
54
|
Ray SK, Mukherjee S. Starring Role of Biomarkers and Anticancer Agents as a Major Driver in Precision Medicine of Cancer Therapy. Curr Mol Med 2023; 23:111-126. [PMID: 34939542 DOI: 10.2174/1566524022666211221152947] [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: 03/19/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022]
Abstract
Precision medicine is the most modern contemporary medicine approach today, based on great amount of data on people's health, individual characteristics, and life circumstances, and employs the most effective ways to prevent and cure diseases. Precision medicine in cancer is the most precise and viable treatment for every cancer patient based on the disease's genetic profile. Precision medicine changes the standard one size fits all medication model, which focuses on average responses to care. Consolidating modern methodologies for streamlining and checking anticancer drugs can have long-term effects on understanding the results. Precision medicine can help explicit anticancer treatments using various drugs and even in discovery, thus becoming the paradigm of future cancer medicine. Cancer biomarkers are significant in precision medicine, and findings of different biomarkers make this field more promising and challenging. Naturally, genetic instability and the collection of extra changes in malignant growth cells are ways cancer cells adapt and survive in a hostile environment, for example, one made by these treatment modalities. Precision medicine centers on recognizing the best treatment for individual patients, dependent on their malignant growth and genetic characterization. This new era of genomics progressively referred to as precision medicine, has ignited a new episode in the relationship between genomics and anticancer drug development.
Collapse
Affiliation(s)
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020. India
| |
Collapse
|
55
|
Zinnah KMA, Munna AN, Seol JW, Park BY, Park SY. An Antidepressant Drug Increased TRAIL Receptor-2 Expression and Sensitized Lung Cancer Cells to TRAIL-induced Apoptosis. Anticancer Agents Med Chem 2023; 23:2225-2236. [PMID: 37859313 PMCID: PMC10788920 DOI: 10.2174/0118715206262252231004110310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND TRAIL has emerged as a promising therapeutic target due to its ability to selectively induce apoptosis in cancer cells while sparing normal cells. Autophagy, a highly regulated cellular recycling mechanism, is known to play a cell survival role by providing a required environment for the cell. Recent studies suggest that autophagy plays a significant role in increasing TRAIL resistance in certain cancer cells. Thus, regulating autophagy in TRAIL-mediated cancer therapy is crucial for its role in cancer treatment. OBJECTIVE Our study explored whether the antidepressant drug desipramine could enhance the ability of TRAIL to kill cancer cells by inhibiting autophagy. METHODS The effect of desipramine on TRAIL sensitivity was examined in various lung cancer cell lines. Cell viability was measured by morphological analysis, trypan blue exclusion, and crystal violet staining. Flow cytometry analysis was carried out to measure apoptosis with annexin V-PI stained cells. Western blotting, rtPCR, and immunocytochemistry were carried out to measure autophagy and death receptor expression. TEM was carried out to detect autophagy inhibition. RESULTS Desipramine treatment increased the TRAIL sensitivity in all lung cancer cell lines. Mechanistically, desipramine treatment induced death receptor expression to increase TRAIL sensitivity. This effect was confirmed when the genetic blockade of DR5 reduced the effect of desipramine in enhanced TRAIL-mediated cell death. Further investigation revealed that desipramine treatment increased the LC3 and p62 levels, indicating the inhibition of lysosomal degradation of autophagy. Notably, TRAIL, in combination with either desipramine or the autophagy inhibitor chloroquine, exhibited enhanced cytotoxicity compared to TRAIL treatment alone. CONCLUSION Our findings revealed the potential of desipramine to induce TRAIL-mediated cell death by autophagy impairment. This discovery suggests its therapeutic potential for inducing TRAIL-mediated cell death by increasing the expression of death receptors, which is caused by impairing autophagy.
Collapse
Affiliation(s)
- Kazi Mohammad Ali Zinnah
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
- Department of Animal and Fish Biotechnology, Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Ali Newaz Munna
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Byung-Yong Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| |
Collapse
|
56
|
Ferah Okkay I, Okkay U, Bayram C, Cicek B, Sezen S, Aydin IC, Mendil AS, Hacimuftuoglu A. Bromelain protects against cisplatin-induced ocular toxicity through mitigating oxidative stress and inflammation. Drug Chem Toxicol 2023; 46:69-76. [PMID: 34894956 DOI: 10.1080/01480545.2021.2011308] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The aim of this study was to investigate the molecular, biochemical, and histopathological effects of bromelain, which has antioxidant and anti-inflammatory properties, against cisplatin-induced ocular toxicity. The groups were designed as (1) Control, (2) Cisplatin (7 mg/kg, intraperitoneally), (3) Cisplatin + Bromelain (50 mg/kg, orally for 14 consecutive days), (4) Cisplatin + Bromelain (100 mg/kg, orally for 14 consecutive days). The activity of total antioxidant capacity (TAC) and total oxidant status (TOS) and levels of reactive oxygen species (ROS), superoxide dismutase (SOD), malondialdehyde (MDA), interleukin-1β (IL-1β), IL-10, nuclear factor kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α) and 8-OHdG were measured in ocular tissue. The mRNA expression of NF-κB and Caspase-3 was also evaluated. Also, ocular sections were evaluated histopathologically. Bromelain demonstrated a dose-dependent protective effect in cisplatin-induced toxicity by regulating oxidative stress, inflammation, and tissue damage. Our results suggested that bromelain may be a potential adjuvant that can protect the eye from cisplatin-induced toxicity.
Collapse
Affiliation(s)
- Irmak Ferah Okkay
- Faculty of Pharmacy, Department of Pharmacology, Ataturk University, Erzurum, Turkey
| | - Ufuk Okkay
- Faculty of Medicine, Department of Medical Pharmacology, Ataturk University, Erzurum, Turkey
| | - Cemil Bayram
- Faculty of Medicine, Department of Medical Pharmacology, Ataturk University, Erzurum, Turkey
| | - Betul Cicek
- Faculty of Medicine, Department of Physiology, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Selma Sezen
- Faculty of Medicine, Department of Medical Pharmacology, Ataturk University, Erzurum, Turkey
| | - Ismail Cagri Aydin
- Faculty of Medicine, Department of Medical Pharmacology, Ataturk University, Erzurum, Turkey.,Faculty of Pharmacy, Department of Pharmacology, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Ali Sefa Mendil
- Faculty of Veterinary Medicine, Department of Pathology, Erciyes University, Kayseri, Turkey
| | - Ahmet Hacimuftuoglu
- Faculty of Medicine, Department of Medical Pharmacology, Ataturk University, Erzurum, Turkey
| |
Collapse
|
57
|
Evaluation of developmental toxicity in zebrafish embryos and antiproliferative potential against human tumor cell lines of new derivatives containing 4-nitrophenyl group. Toxicol Appl Pharmacol 2023; 458:116325. [PMID: 36436567 DOI: 10.1016/j.taap.2022.116325] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
The aim of the studies was to evaluate the antiproliferative potential against human tumor cell lines of newly synthetized derivatives containing 4-nitrophenyl group, as well as its impact on developmental toxicity in zebrafish model. We selected 1-(4-nitrobenzoyl)-4-ethylsemicarbazide (APS-1) and 1-[(4-nitrophenyl)acetyl]-4-hexyl-thiosemicarbazide (APS-18) for research. The antiproliferative properties of semicarbazide derivatives were assessed against human cancer cell lines derived from hepatocellular adenocarcinoma (HepG2), renal cell carcinoma (769-P), non-small cell lung cancer (NCI-H1563) and glioblastoma multiforme (LN229) in comparison to the physiological human embryonic kidney (HEK-293) cell line. The influence of the tested substances on the cell cycle and apoptosis was also evaluated. Fish embryo acute toxicity test (FET) was performed based on OECD Guidelines (Test No. 236), and was carried out for the first 5 days post fertilization. The following concentrations of APS-1 and APS-18 were tested: 125-2000 μM and 0.125-1000 μM, respectively. The presented studies on the antiproliferative properties of the new semicarbazide derivatives showed that the compounds APS-1 and APS-18 reduce the viability of human tumor lines. Particularly noteworthy is the strong and selective antiproliferative activity of APS-18 against all neoplastic cell lines, in particular against glioblastoma. Against this tumor line, the compound APS-1 showed an effective inhibitory effect. In the FET we noted that the direct exposure of zebrafish embryos to APS-1 and APS-18 in used range of concentration did not cause morphological abnormalities, including cardiotoxicity. On basis of obtained outcomes it could be concluded that APS-1 and APS-18 may constitute models for further research, design and synthesis of new, safer drugs with more favorable anticancer properties.
Collapse
|
58
|
Hawash M, Qaoud MT, Jaradat N, Abdallah S, Issa S, Adnan N, Hoshya M, Sobuh S, Hawash Z. Anticancer Activity of Thiophene Carboxamide Derivatives as CA-4 Biomimetics: Synthesis, Biological Potency, 3D Spheroid Model, and Molecular Dynamics Simulation. Biomimetics (Basel) 2022; 7:247. [PMID: 36546947 PMCID: PMC9775471 DOI: 10.3390/biomimetics7040247] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The present study aimed to synthesize thiophene carboxamide derivatives, which are considered biomimetics of the anticancer medication Combretastatin A-4 (CA-4), and compare the similarity in the polar surface area (PSA) between the novel series and CA-4. Our results showed that the PSA of the most synthesized structures was biomimetic to CA-4, and similar chemical and biological properties were observed against Hep3B cancer cell line. Among the synthesized series 2b and 2e compounds were the most active molecules on Hep3B (IC50 = 5.46 and 12.58 µM, respectively). The 3D results revealed that both 2b and 2e structures confuse the surface of Hep3B cancer cell lines' spheroid formation and force these cells to aggregate into a globular-shaped spheroid. The 2b and 2e showed a comparable interaction pattern to that observed for CA-4 and colchicine within the tubulin-colchicine-binding pocket. The thiophene ring, due to holding a high aromaticity character, participated critically in that observed interaction profile and showed additional advanced interactions over CA-4. The 2b and 2e tubulin complexes showed optimal dynamics trajectories within a time scale of 100 ns at 300 K temperature, which asserts their high stability and compactness. Together, these findings revealed the biomimetic role of 2b and 2e compounds in CA-4 in preventing cancer progression.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine
| | - Mohammed T. Qaoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Etiler, 06330 Ankara, Turkey
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine
| | - Samer Abdallah
- Department of Biology & Biotechnology, Faculty of Science, An-Najah National University, Nablus 00970, Palestine
| | - Shahd Issa
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine
| | - Nawal Adnan
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine
| | - Marah Hoshya
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine
| | - Shorooq Sobuh
- Department of Biomedical Sciences, Physiology, Pharmacology & Toxicology Division, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine
| | - Zafer Hawash
- Department of Physics, Faculty of Science, Birzeit University, Birzeit, Ramallah 71939, Palestine
| |
Collapse
|
59
|
Reversal of multidrug resistance by Fissistigma latifolium–derived chalconoid 2-hydroxy-4,5,6-trimethoxydihydrochalcone in cancer cell lines overexpressing human P-glycoprotein. Biomed Pharmacother 2022; 156:113832. [DOI: 10.1016/j.biopha.2022.113832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/24/2022] [Accepted: 10/05/2022] [Indexed: 11/23/2022] Open
|
60
|
Sabet Z, Vagiannis D, Budagaga Y, Zhang Y, Novotná E, Hanke I, Rozkoš T, Hofman J. Talazoparib Does Not Interact with ABCB1 Transporter or Cytochrome P450s, but Modulates Multidrug Resistance Mediated by ABCC1 and ABCG2: An in Vitro and Ex Vivo Study. Int J Mol Sci 2022; 23:ijms232214338. [PMID: 36430819 PMCID: PMC9697930 DOI: 10.3390/ijms232214338] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Talazoparib (Talzenna) is a novel poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitor that is clinically used for the therapy of breast cancer. Furthermore, the drug has shown antitumor activity against different cancer types, including non-small cell lung cancer (NSCLC). In this work, we investigated the possible inhibitory interactions of talazoparib toward selected ATP-binding cassette (ABC) drug efflux transporters and cytochrome P450 biotransformation enzymes (CYPs) and evaluated its position in multidrug resistance (MDR). In accumulation studies, talazoparib interacted with the ABCC1 and ABCG2 transporters, but there were no significant effects on ABCB1. Furthermore, incubation assays revealed a negligible capacity of the tested drug to inhibit clinically relevant CYPs. In in vitro drug combination experiments, talazoparib synergistically reversed daunorubicin and mitoxantrone resistance in cells with ABCC1 and ABCG2 expression, respectively. Importantly, the position of an effective MDR modulator was further confirmed in drug combinations performed in ex vivo NSCLC patients-derived explants, whereas the possible victim role was refuted in comparative proliferation experiments. In addition, talazoparib had no significant effects on the mRNA-level expressions of MDR-related ABC transporters in the MCF-7 cellular model. In summary, our study presents a comprehensive overview on the pharmacokinetic drug-drug interactions (DDI) profile of talazoparib. Moreover, we introduced talazoparib as an efficient MDR antagonist.
Collapse
Affiliation(s)
- Ziba Sabet
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Dimitrios Vagiannis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Youssif Budagaga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Yu Zhang
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Eva Novotná
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Ivo Hanke
- Department of Cardiac Surgery, Faculty of Medicine, Charles University in Hradec Králové and University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Králové, Czech Republic
| | - Tomáš Rozkoš
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine and University Hospital in Hradec Králové, Czech Republic, Sokolská 581, 500 05 Hradec Králové, Czech Republic
| | - Jakub Hofman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
- Correspondence: ; Tel.: +420-495-067-593
| |
Collapse
|
61
|
TNBC Therapeutics Based on Combination of Fusarochromanone with EGFR Inhibitors. Biomedicines 2022; 10:biomedicines10112906. [PMID: 36428475 PMCID: PMC9687139 DOI: 10.3390/biomedicines10112906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
Fusarochromanone is an experimental drug with unique and potent anti-cancer activity. Current cancer therapies often incorporate a combination of drugs to increase efficacy and decrease the development of drug resistance. In this study, we used drug combinations and cellular phenotypic screens to address important questions about FC101's mode of action and its potential therapeutic synergies in triple negative breast cancer (TNBC). We hypothesized that FC101's activity against TNBC is similar to the mTOR inhibitor, everolimus, because FC101 downregulates the phosphorylation of two mTOR substrates, S6K and S6. Since everolimus synergistically enhances the anti-cancer activities of two known EGFR inhibitors (erlotinib or lapatinib) in TNBC, we performed analogous studies with FC101. Phenotypic cellular assays helped assess whether FC101 acts similarly to everolimus, in both single and combination treatments with the two inhibitors. FC101 outperformed all other single treatments in both cell proliferation and viability assays. However, unlike everolimus, FC101 produced a sustained decrease in cell viability in drug washout studies. None of the other drugs were able to maintain comparable effects upon removal. Although we observed slightly additive effects when the TNBC cells were treated with FC101 and the two EGFR inhibitors, those effects were not truly synergistic in the manner displayed with everolimus.
Collapse
|
62
|
Sim TM. Nanoparticle-assisted targeting of the tumour microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
63
|
Koopaei NN, Shademani M, Yazdi NS, Tahmasvand R, Dehbid M, Koopaei MN, Azizian H, Mousavi Z, Almasirad A, Salimi M. Design and synthesis of novel ureido and thioureido conjugated hydrazone derivatives with potent anticancer activity. BMC Chem 2022; 16:81. [PMID: 36320042 PMCID: PMC9624014 DOI: 10.1186/s13065-022-00873-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Background Compounds possessing urea/thiourea moiety have a wide range of biological properties including anticancer activity. On the other hand, taking advantage of the low toxicity and structural diversity of hydrazone derivatives, they are presently being considered for designing chemical compounds with hydrazone moiety in the field of cancer treatment. With this in mind, a series of novel ureido/thioureido derivatives possessing a hydrazone moiety bearing nitro and chloro substituents (4a–4i) have been designed, synthesized, characterized and evaluated for their in vitro cytotoxic effect on HT-29 human colon carcinoma and HepG2 hepatocarcinoma cell lines. Results Two compounds (4c and 4e) having the chloro phenylurea group hybridized with phenyl hydrazone bearing nitro or chloro moieties demonstrated potent anticancer effect with the IC50 values between 2.2 and 4.8 µM at 72 h. The mechanism of action of compound 4c was revealed in hepatocellular carcinoma cells as an inducer of apoptosis in a caspase-independent pathway. Conclusion Taken together, the current work presented compound 4c as a potential lead compound in developing future hepatocellular carcinoma chemotherapy drugs. Methods The compounds were synthesized and then characterized by physical and spectral data (FT-IR, 1H-NMR, 13C-NMR, Mass). The anticancer activity was assessed using MTT assay, flowcytometry, annexin-V, DAPI staining and Western blot analysis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13065-022-00873-3.
Collapse
Affiliation(s)
- Nasrin Nassiri Koopaei
- grid.411463.50000 0001 0706 2472Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, P.O. Box 1941933111, Tehran, Iran
| | - Mehrasa Shademani
- grid.420169.80000 0000 9562 2611Department of Physiology and Pharmacology, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran ,grid.411463.50000 0001 0706 2472Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nasrin Shirzad Yazdi
- grid.411463.50000 0001 0706 2472Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, P.O. Box 1941933111, Tehran, Iran ,grid.412571.40000 0000 8819 4698Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raheleh Tahmasvand
- grid.420169.80000 0000 9562 2611Department of Physiology and Pharmacology, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Mina Dehbid
- grid.411872.90000 0001 2087 2250Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mansur Nassiri Koopaei
- grid.411705.60000 0001 0166 0922Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design & Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Azizian
- grid.411746.10000 0004 4911 7066Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Science, Tehran, Iran
| | - Zahra Mousavi
- grid.411463.50000 0001 0706 2472Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Almasirad
- grid.411463.50000 0001 0706 2472Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, P.O. Box 1941933111, Tehran, Iran
| | - Mona Salimi
- grid.420169.80000 0000 9562 2611Department of Physiology and Pharmacology, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| |
Collapse
|
64
|
Alkhalifah MA, Yar M, Bayach I, Sheikh NS, Ayub K. Covalent Organic Framework (C 6N 6) as a Drug Delivery Platform for Fluorouracil to Treat Cancerous Cells: A DFT Study. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7425. [PMID: 36363017 PMCID: PMC9654209 DOI: 10.3390/ma15217425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 06/16/2023]
Abstract
Continuous studies are being carried out to explore new methods and carrier surfaces for target drug delivery. Herein, we report the covalent triazine framework C6N6 as a drug delivery carrier for fluorouracil (FU) and nitrosourea (NU) anti-cancer drugs. FU and NU are physiosorbed on C6N6 with adsorption energies of -28.14 kcal/mol and -27.54 kcal/mol, respectively. The outcomes of the non-covalent index (NCI) and quantum theory of atoms in molecules (QTAIM) analyses reveal that the FU@C6N6 and NU@C6N6 complexes were stabilized through van der Waals interactions. Natural bond order (NBO) and electron density difference (EDD) analyses show an appreciable charge transfer from the drug and carrier. The FU@C6N6 complex had a higher charge transfer (-0.16 e-) compared to the NU@C6N6 complex (-0.02 e-). Frontier molecular orbital (FMO) analysis reveals that the adsorption of FU on C6N6 caused a more pronounced decrease in the HOMO-LUMO gap (EH-L) compared to that of NU. The results of the FMO analysis are consistent with the NBO and EDD analyses. The drug release mechanism was studied through dipole moments and pH effects. The highest decrease in adsorption energy was observed for the FU@C6N6 complex in an acidic medium, which indicates that FU can easily be off-loaded from the carrier (C6N6) to a target site because the cancerous cells have a low pH compared to a normal cell. Thus, it may be concluded that C6N6 possesses the therapeutic potential to act as a nanocarrier for FU to treat cancer. Furthermore, the current study will also provide motivation to the scientific community to explore new surfaces for drug delivery applications.
Collapse
Affiliation(s)
- Mohammed A. Alkhalifah
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Muhammad Yar
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
| | - Imene Bayach
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Nadeem S. Sheikh
- Chemical Sciences, Faculty of Science, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong BE1410, Brunei
| | - Khurshid Ayub
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
| |
Collapse
|
65
|
Akash S, Kumer A, Rahman MM, Emran TB, Sharma R, Singla RK, Alhumaydhi FA, Khandaker MU, Park MN, Idris AM, Wilairatana P, Kim B. Development of new bioactive molecules to treat breast and lung cancer with natural myricetin and its derivatives: A computational and SAR approach. Front Cell Infect Microbiol 2022; 12:952297. [PMID: 36237438 PMCID: PMC9551266 DOI: 10.3389/fcimb.2022.952297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Each biopharmaceutical research and new drug development investigation is targeted at discovering novel and potent medications for managing specific ailments. Thus, to discover and develop new potent medications, it should be performed sequentially or step by step. This is because drug development is a lengthy and risky work that requires significant money, resources, and labor. Breast and lung cancer contributes to the death of millions of people throughout the world each year, according to the report of the World Health Organization, and has been a public threat worldwide, although the global medical sector is developed and updated day by day. However, no proper treatment has been found until now. Therefore, this research has been conducted to find a new bioactive molecule to treat breast and lung cancer-such as natural myricetin and its derivatives-by using the latest and most authentic computer-aided drug-design approaches. At the beginning of this study, the biological pass prediction spectrum was calculated to select the target protein. It is noted that the probability of active (Pa) score is better in the antineoplastic (Pa: 0.788-0.938) in comparison with antiviral (Pa: 0.236-0.343), antibacterial (Pa: 0.274-0.421), and antifungal (Pa: 0.226-0.508). Thus, cancerous proteins, such as in breast and lung cancer, were picked up, and the computational investigation was continued. Furthermore, the docking score was found to be -7.3 to -10.4 kcal/mol for breast cancer (standard epirubicin hydrochloride, -8.3 kcal/mol), whereas for lung cancer, the score was -8.2 to -9.6 kcal/mol (standard carboplatin, -5.5 kcal/mol). The docking score is the primary concern, revealing that myricetin derivatives have better docking scores than standard chemotherapeutic agents epirubicin hydrochloride and carboplatin. Finally, drug-likeness, ADME, and toxicity prediction were fulfilled in this investigation, and it is noted that all the derivatives were highly soluble in a water medium, whereas they were totally free from AMES toxicity, hepatotoxicity, and skin sensitization, excluding only ligands 1 and 7. Thus, we proposed that the natural myricetin derivatives could be a better inhibitor for treating breast and lung cancer.
Collapse
Affiliation(s)
- Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Ajoy Kumer
- Laboratory of Computational Research for Drug Design and Material Science, Department of Chemistry, European University of Bangladesh, Dhaka, Bangladesh
| | - Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - Moon Nyeo Park
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Abubakr M. Idris
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Bonglee Kim
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
66
|
Nanoparticles Design for Theranostic Approach in Cancer Disease. Cancers (Basel) 2022; 14:cancers14194654. [PMID: 36230578 PMCID: PMC9564040 DOI: 10.3390/cancers14194654] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Presently, there are no conclusive treatments for many types of cancer, mainly due to the advanced phase of the disease at the time of diagnosis and to the side effects of existing therapies. Present diagnostic and therapeutic procedures need to be improved to supply early detection abilities and perform a more specific therapy with reduced systemic toxicity. In this review, improvements in nanotechnology allowing the design of multifunctional nanoparticles for cancer detection, therapy, and monitoring are reported. Nanoparticles, thanks to the nanomaterials they are made of, can be used as contrast agents for various diagnostic techniques such as MRI, optical imaging, and photoacoustic imaging. Furthermore, when used as drug carriers, they can accumulate in tumor tissues through the passive or/and active targeting, protect encapsulated drugs from degradation, raise tumor exposure to chemotherapeutic agents improving treatment effects. In addition, nanocarriers can simultaneously deliver more than one therapeutic agent enhancing the effectiveness of therapy and can co-deliver imaging and therapy agents to provide integration of diagnostics, therapy, and follow-up. Furthermore, the use of nanocarriers allows to use different therapeutic approaches, such as chemotherapy and hyperthermia to exploit synergistic effects. Theranostic approach to diagnose and treat cancer show a great potential to improve human health, however, despite technological advances in this field, the transfer into clinical practice is still a long way off.
Collapse
|
67
|
Christodoulou P, Boutsikos P, Neophytou CM, Kyriakou TC, Christodoulou MI, Papageorgis P, Stephanou A, Patrikios I. Amygdalin as a chemoprotective agent in co-treatment with cisplatin. Front Pharmacol 2022; 13:1013692. [PMID: 36204233 PMCID: PMC9531591 DOI: 10.3389/fphar.2022.1013692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Amygdalin is a naturally occurring glycoside used in traditional Chinese medicine and is known to have anti-cancer properties. Even though the anti-cancer properties of amygdalin are well known, its effect on normal cells has not been thoroughly investigated. The aim of the present study was to investigate a possible chemo-protective role of amygdalin against the cytotoxic effects of chemotherapy for normal human cells. Specifically, it was tested in combination with a strong chemotherapeutic drug cisplatin. Human non-tumorigenic MCF12F epithelial cell line, human fibroblasts cells, human breast cancer MCF7 and MDA-MB-231 cells were treated with cisplatin in a dose- and time-depended manner in the absence or presence of amygdalin. When MCF12F cells and fibroblasts underwent pre-treatment with amygdalin followed by cisplatin treatment (24 h amygdalin + 24 h cisplatin), the cell viability was increased (22%, p < 0.001) as indicated using MTT assay. As attested by flow cytometry, combination treatment was associated with decreased the percentage of late apoptotic cells compared with monotherapy (fold-change of decrease = 1.6 and 4.5 for 15 and 20 μΜ, respectively). Also, the proteins expression of PUMA, p53, phospho-p53 and Bax decreased, when a combination treatment was used vs. cisplatin alone, while the proapoptotic proteins Bcl-2 and Bcl-xL exhibited an increased tendency in the presence of amygdalin. Moreover, the levels of pro-apoptotic genes PUMA, p53, and BAX mRNA were significantly downregulated (∼83%, ∼66%, and ∼44%, respectively) vs. cisplatin alone, while the mRNA levels of anti-apoptotic genes BCl-2 and Bcl-XL were upregulated (∼44.5% and ∼51%, respectively), vs. cisplatin alone after 24 h of combination treatment. The study on the Combination index (CI) assay indicated that amygdalin could be possibly considered as an antagonist to cisplatin (2.2 and 2.3) for MCF12F and fibroblast cells, respectively. In contrast, for the breast cancer MCF7 and MDA-MB-231 cells, amygdalin and cisplatin indicated a synergistic effect (0.8 and 0.65), respectively. Our present findings suggest that amygdalin has chemo-modulatory effect when used in co-treatment with cisplatin and is able to protect normal breast cells as well as the fibroblasts during chemotherapy treatment, indicating a strong selective chemoprotective ability and may contribute to a better quality of life for cancer patients.
Collapse
Affiliation(s)
- Panayiota Christodoulou
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Tumor Immunology and Biomarkers Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
- *Correspondence: Panayiota Christodoulou,
| | | | - Christiana M. Neophytou
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
| | - Theodora-Christina Kyriakou
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
| | - Maria-Ioanna Christodoulou
- Tumor Immunology and Biomarkers Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
| | - Panagiotis Papageorgis
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
| | | | | |
Collapse
|
68
|
Hadi H, Safari R, Shamlouei HR. Synthesis and experimental/theoretical evaluation of β-CD/MTX nanostructure for use in targeted drug delivery systems. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02459-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
69
|
Anti-lung cancer properties of cyanobacterial bioactive compounds. Arch Microbiol 2022; 204:603. [PMID: 36063223 DOI: 10.1007/s00203-022-03194-0] [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: 02/15/2022] [Revised: 08/07/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022]
Abstract
Lung cancer, the most prevalent gender-independent tumor entity in both men and women, is among the leading cause of cancer-related deaths worldwide. Despite decades of effort in developing improved therapeutic strategies including immunotherapies and novel chemotherapeutic agents, only modest improvements in outcome and long-term survival of lung cancer patients have been achieved. Therefore, exploring new and exceptional sources for bioactive compounds that might serve as anti-cancer agents might be the key to improving lung cancer therapy. On account of diverse forms, cyanobacteria might serve as a potential source for compounds with potential therapeutic applicability against malignant disorders, including cancer. The assorted arrays of metabolic mechanisms synthesize a plethora of bioactive compounds with immense biological potential. These compounds have been proven to be effective against various cancer cell lines and xenograft animal models. The present review provides an overview of the most promising cyanobacteria-derived bioactive compounds proven to exhibit anti-cancer properties in in-vitro and in-vivo studies and highlights their applicability as potential therapeutic agents with a focus on their anti-lung cancer properties.
Collapse
|
70
|
Lozon L, Saleh E, Menon V, Ramadan WS, Amin A, El-Awady R. Effect of safranal on the response of cancer cells to topoisomerase I inhibitors: Does sequence matter? Front Pharmacol 2022; 13:938471. [PMID: 36120345 PMCID: PMC9479137 DOI: 10.3389/fphar.2022.938471] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022] Open
Abstract
Lung and colorectal cancers are among the leading causes of death from cancer worldwide. Although topotecan (TPT), a topoisomerase1 inhibitor, is a first- and second-line drug for lung and colon cancers, the development of drug resistance and toxicity still remain as a major obstacle to chemotherapeutic success. Accumulating evidence indicates increased efficacy and reduced toxicity of chemotherapeutic agents upon combining them with natural products. We aimed to investigate the possible interaction of safranal (SAF), a natural compound obtained from Crocus sativus stigma, with TPT when used in different sequences in colon and lung cancer cell lines. The growth inhibitory effect of the proposed combination given in different sequences was assessed using the colony formation assay. The comet assay, cell cycle distribution, Annexin-V staining, and expression of proteins involved in DNA damage/repair were utilized to understand the mechanism underlying the effect of the combination. SAF enhanced the growth inhibitory effects of TPT particularly when it was added to the cells prior to TPT. This combination increased the double-strand break induction and dysregulated the DNA repair machinery, particularly the tyrosyl-DNA phosphodiesterase 1 enzyme. In addition, the SAF + TPT combination increased the fraction of cells arrested at the G2/M checkpoint as well as enhanced the induction of apoptosis. The current study highlights the status of SAF as a natural product sensitizing the lung and colon cancer cells to the cytotoxic effects of the anticancer drug TPT. In addition, it emphasizes the importance of sequence-dependent interaction which can affect the overall outcome.
Collapse
Affiliation(s)
- Lama Lozon
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ekram Saleh
- Clinical Biochemistry and Molecular Biology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Giza, Egypt
| | - Varsha Menon
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Wafaa S. Ramadan
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Amr Amin
- Department of Biology, College of Science, UAE University, Al Ain, United Arab Emirates
| | - Raafat El-Awady
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
71
|
Bhattacharjee R, Dey T, Kumar L, Kar S, Sarkar R, Ghorai M, Malik S, Jha NK, Vellingiri B, Kesari KK, Pérez de la Lastra JM, Dey A. Cellular landscaping of cisplatin resistance in cervical cancer. Biomed Pharmacother 2022; 153:113345. [PMID: 35810692 DOI: 10.1016/j.biopha.2022.113345] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022] Open
Abstract
Cervical cancer (CC) caused by human papillomavirus (HPV) is one of the largest causes of malignancies in women worldwide. Cisplatin is one of the widely used drugs for the treatment of CC is rendered ineffective owing to drug resistance. This review highlights the cause of resistance and the mechanism of cisplatin resistance cells in CC to develop therapeutic ventures and strategies that could be utilized to overcome the aforementioned issue. These strategies would include the application of nanocarries, miRNA, CRIPSR/Cas system, and chemotherapeutics in synergy with cisplatin to not only overcome the issues of drug resistance but also enhance its anti-cancer efficiency. Moreover, we have also discussed the signaling network of cisplatin resistance cells in CC that would provide insights to develop therapeutic target sites and inhibitors. Furthermore, we have discussed the role of CC metabolism on cisplatin resistance cells and the physical and biological factors affecting the tumor microenvironments.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Tanima Dey
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, Kerala, India
| | - Sulagna Kar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Ritayan Sarkar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Mimosa Ghorai
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand 834001, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India.
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641-046, India
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo 00076, Finland; Department of Bio-products and Bio-systems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - José M Pérez de la Lastra
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), Avda. Astrofísico Francisco Sánchez, 3, 38206 San Cristóbal de la Laguna (Santa Cruz de Tenerife), Spain.
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|
72
|
Chiu WJ, Lin CS, Lin SR, Chen TH, Wu CJ, Busa P, Long H, Chen CC, Tseng FJ, Fu YS, Weng CF. Diterpene promptly executes a non-canonical autophagic cell death in doxorubicin-resistant lung cancer. Biomed Pharmacother 2022; 153:113443. [DOI: 10.1016/j.biopha.2022.113443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022] Open
|
73
|
Investigation of the multi-targeted protection potential of tannic acid against doxorubicin-induced kidney damage in rats. Chem Biol Interact 2022; 365:110111. [PMID: 35987278 DOI: 10.1016/j.cbi.2022.110111] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/20/2022]
Abstract
Doxorubicin (DOX) is an antitumor drug that is powerful but can cause worse outcomes, including nephrotoxicity, and therefore has limited clinical use. Therefore, it is necessary to identify safer agents that can minimize the damage caused by the drug without shifting the treatment performance, in addition to clarifying the underlying mechanisms of DOX-induced aberrant in vivo renal activation. In this study, we tested the prophylactic capacity and mechanisms of action of tannic acid (TA) against DOX-mediated kidney damage in rats and evaluated the nephrotoxic activity of DOX when used with TA. Rats were treated during the two weeks with cumulative (18 mg/kg with six different injections) DOX, daily TA (50 mg/kg), and the DOX + TA combination. Changes in major metabolites and components involved in antioxidant metabolism were evaluated in the kidney tissues of all animals. Further, the gene expression levels of regulatory factors that have critical importance in cell metabolism, inflammation, and apoptosis were investigated. Both biochemical and molecular examinations showed that TA improved DOX-induced dysregulations at both protein and gene levels in the kidneys. Increased lipid peroxidation and decreased glutathione levels were reversed. Consistent with oxidative stress marker metabolites, suppressed antioxidant enzyme activities and transcript levels of antioxidant system members were restored. Of note, combination treatment with TA could overcome doxorubicin-induced gene expressions markedly altered by DOX, suggesting that nephroprotection conferred by TA involved the remodeling of stress resistance, cell metabolism, inflammation, and apoptosis. Collectively, the present in vivo study suggests that TA could be used as a multitarget and effective agent for the mitigation of doxorubicin-induced nephrotoxicity without changing the therapeutic efficacy of the drug.
Collapse
|
74
|
Khalaji A, Haddad S, Yazdani Y, Moslemi M, Alizadeh L, Baradaran B. A bioinformatics-based study on the Cisplatin-resistant lung cancer cells; what are the orchestrators of this phenom? Gene X 2022; 834:146668. [PMID: 35690284 DOI: 10.1016/j.gene.2022.146668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 11/04/2022] Open
Abstract
Lung cancer represents a significant global health issue and is among the central causes of mortality and morbidity around the world. Unfortunately, the majority of lung cancer patients acquire drug resistant to chemotherapy either intrinsically or acquired after Cisplatin treatment. It is indicated that increasing or decreasing the expression of particular genes can affect chemotherapeutic sensitivity or resistance. As a result, gaining a deeper knowledge of the changed expression of genes implicated in lung cancer drug resistance, as well as developing novel therapeutic techniques, are critical targets for continued advancement in lung cancer treatment. In the present study, we aimed to find key regulatory genes in the progression of Cisplatin resistance in A-549 lung cancer cells. In this regard, microarray dataset of Cisplatin-resistant and Cisplatin-sensitive was retrieved from the Gene Expression Omnibus (GEO) with accession number of GSE108214. Then, differentially expressed genes (DEGs) between sensitive and resistant lung cancer cells were obtained by using R software v4.0.2 and related packages. We recognized CEACAM1, DGKA, ARHGEF4, and THSD4 are involved in the drug resistance. Experimentally, Cisplatin-resistant A-549 cells were developed and analyzed by MTT assay. Besides, the expression of candidate genes were analyzed in these cells compared to Cisplatin-sensitive A-549 cells by qRT-PCR. The findings presented that the expression of CEACAM1, DGKA, ARHGEF4, and THSD4 was altered following the induction of Cisplatin resistance in A549 cells.
Collapse
Affiliation(s)
- Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Haddad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Yazdani
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Emam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Moslemi
- Department of Internal Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Leila Alizadeh
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
75
|
Al-Jumaili MHA, Hamad AA, Hashem HE, Hussein AD, Muhaidi MJ, Ahmed MA, ALBANAA AHA, Siddique F, Bakr EA. Comprehensive Review on the Bis–heterocyclic Compounds and their Anticancer Efficacy. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
76
|
Ji X, Cheng K, Gao C, Xie H, Zhu R, Luo J. HS3ST1 Promotes Non-Small-Cell Lung Cancer Progression by Targeting the SPOP/FADD/NF- κB Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5509346. [PMID: 35909476 PMCID: PMC9325619 DOI: 10.1155/2022/5509346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 01/04/2023]
Abstract
Heparan sulfate proteoglycan is a key component of cell microenvironment and plays an important role in cell-cell interaction, adhesion, migration, and signal transduction. Heparan sulfate 3-O-sulfotransferase 1 (HS3ST1) is a metabolic-related gene of HS. The present study was aimed at exploring the role of HS3ST1 in the progress of non-small-cell lung cancer (NSCLC). Our results illustrated that HS3ST1 promoted the malignant behaviors of NSCLC cells both in vitro and in vivo. HS3ST1 was found to inhibit spot-type zinc finger protein (SPOP) expression, which might inhibit the NF-κB pathway activation through mediating the degradation of Fas-associated death domain protein (FADD). By analyzing NSCLC patient samples, we also found increased HS3ST1 expression and decreased SPOP expression in tumor tissues in contrast with those in adjoining normal tissues. In conclusion, HS3ST1 promotes NSCLC tumorigenesis by regulating SPOP/FADD/NF-κB pathway.
Collapse
Affiliation(s)
- Xianxiu Ji
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Kebin Cheng
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Caixia Gao
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Ren Zhu
- Department of Medical Administration, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Jie Luo
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
77
|
Maji M, Kivale P, Ghosh M. A novel therapy to combat non-small cell lung carcinoma (A549) using platinum (IV) and barium titanate conjugate. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
78
|
Hawash M, Jaradat N, Eid AM, Abubaker A, Mufleh O, Al-Hroub Q, Sobuh S. Synthesis of novel isoxazole-carboxamide derivatives as promising agents for melanoma and targeted nano-emulgel conjugate for improved cellular permeability. BMC Chem 2022; 16:47. [PMID: 35751124 PMCID: PMC9229817 DOI: 10.1186/s13065-022-00839-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cancer is one of the most dangerous and widespread diseases in the world today and it has risen to the position of the leading cause of death around the globe in the last few decades. Due to the inherent resistance of many types of cancer to conventional radiotherapy and chemotherapy, it is vital to develop innovative anticancer medications. Recently, a strategy based on nanotechnology has been used to improve the effectiveness of both old and new cancer drugs. OBJECTIVES The present study aimed to design and synthesize a series of phenyl-isoxazole-Carboxamide derivatives, evaluate their anticancer properties, and improve the permeability of potent compounds into cancer cells by using a nano-emulgel strategy. METHODS The coupling reaction of aniline derivatives and isoxazole-Carboxylic acid was used to synthesize a series of isoxazole-Carboxamide derivatives. IR, HRMS, 1H-NMR, and 13C-NMR spectroscopy techniques, characterized all the synthesized compounds. The in-vitro cytotoxic evaluation was performed by using the MTS assay against seven cancer cell lines, including hepatocellular carcinoma (Hep3B and HepG2), cervical adenocarcinoma (HeLa), breast carcinoma (MCF-7), melanoma (B16F1), colorectal adenocarcinoma (Caco-2), and colon adenocarcinoma (Colo205), as well as human hepatic stellate (LX-2) in addition to the normal cell line (Hek293T). A nano-emulgel was developed for the most potent compound, using a self-emulsifying technique. RESULTS All synthesized compounds were found to have potent to moderate activities against B16F1, Colo205, and HepG2 cancer cell lines. The results revealed that the 2a compound has broad spectrum activity against B16F1, Colo205, HepG2, and HeLa cancer cell lines with an IC50 range of 7.55-40.85 µM. Moreover, compound 2e was the most active compound against B16F1 with an IC50 of 0.079 µM compared with Dox (IC50 = 0.056 µM). Nanoemulgel was used to increase the potency of the 2e molecule against this cancer cell line, and the IC50 was reduced to 0.039 µM. The antifibrotic activities were investigated against the LX-2 cell line, and it was found that our synthesized molecules showed better antifibrotic activities at 1 µM than 5-FU, and the cell viability values were 67 and 95%, respectively. CONCLUSION This study suggests that a 2e nano-formalized compound is a potential and promising anti-melanoma agent.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970, Palestine.
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970, Palestine
| | - Ahmad M Eid
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970, Palestine
| | - Ahmad Abubaker
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970, Palestine
| | - Ola Mufleh
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970, Palestine
| | - Qusay Al-Hroub
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970, Palestine
| | - Shorooq Sobuh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970, Palestine
| |
Collapse
|
79
|
Krasteva N, Georgieva M. Promising Therapeutic Strategies for Colorectal Cancer Treatment Based on Nanomaterials. Pharmaceutics 2022; 14:pharmaceutics14061213. [PMID: 35745786 PMCID: PMC9227901 DOI: 10.3390/pharmaceutics14061213] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a global health problem responsible for 10% of all cancer incidences and 9.4% of all cancer deaths worldwide. The number of new cases increases per annum, whereas the lack of effective therapies highlights the need for novel therapeutic approaches. Conventional treatment methods, such as surgery, chemotherapy and radiotherapy, are widely applied in oncology practice. Their therapeutic success is little, and therefore, the search for novel technologies is ongoing. Many efforts have focused recently on the development of safe and efficient cancer nanomedicines. Nanoparticles are among them. They are uniquewith their properties on a nanoscale and hold the potential to exploit intrinsic metabolic differences between cancer and healthy cells. This feature allows them to induce high levels of toxicity in cancer cells with little damage to the surrounding healthy tissues. Graphene oxide is a promising 2D material found to play an important role in cancer treatments through several strategies: direct killing and chemosensitization, drug and gene delivery, and phototherapy. Several new treatment approaches based on nanoparticles, particularly graphene oxide, are currently under research in clinical trials, and some have already been approved. Here, we provide an update on the recent advances in nanomaterials-based CRC-targeted therapy, with special attention to graphene oxide nanomaterials. We summarise the epidemiology, carcinogenesis, stages of the CRCs, and current nanomaterials-based therapeutic approaches for its treatment.
Collapse
Affiliation(s)
- Natalia Krasteva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., bl. 21, 1113 Sofia, Bulgaria
- Correspondence: (N.K.); (M.G.); Tel.: +359-889-577-074 (N.K.); +359-896-833-604 (M.G.)
| | - Milena Georgieva
- Institute of Molecular Biology “Acad. R. Tsanev”, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., bl. 21, 1113 Sofia, Bulgaria
- Correspondence: (N.K.); (M.G.); Tel.: +359-889-577-074 (N.K.); +359-896-833-604 (M.G.)
| |
Collapse
|
80
|
El-Banna MA, Hendawy OM, El-Nekeety AA, Abdel-Wahhab MA. Efficacy of ginsenoside Rg3 nanoparticles against Ehrlich solid tumor growth in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43814-43825. [PMID: 35118592 DOI: 10.1007/s11356-022-19019-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Solid tumors are fairly common and face many clinical difficulties since they are hardly surgically resectable and broadly do not respond to radiation and chemotherapy. The current study aimed to fabricate ginsenoside Rg3 nanoparticles (Rg3-NPs) and evaluate their antitumor effect against Ehrlich solid tumors (EST) in mice. Rg3-NPs were fabricated using whey protein isolates (WPI), maltodextrin (MD), and gum Arabic (GA). EST was developed by the injection of mice with Ehrlich ascites cells (2.5 × 106). The mice were divided into a control group, EST group, and the EST groups that were treated orally 2 weeks for with normal Rg3 (3 mg/kg b.w.), Rg3-NPs at a low dose (3 mg/kg b.w.), and Rg3-NPs at a high dose (6 mg/kg b.w.). Serum and solid tumors were collected for different assays. The results revealed that synthesized Rg3-NPs showed a spherical shape with an average particle size of 20 nm and zeta potential of -5.58 mV. The in vivo study revealed that EST mice showed a significant increase in AFP, Casp3, TNF-α, MMP-9, VEGF, MDA, and DNA damage accompanied by a significant decrease in SOD and GPx. Treatment with Rg3 or Rg3-NPs decreased the tumor weight and size and induced a significant improvement in all the biochemical parameters. Rg3-NPs were more effective than Rg3, and the improvement was dose-dependent. It could be concluded that fabrication of Rg3-NPs enhanced the protective effect against EST development which may be due to the synergistic effect of Rg3 and MD, GA, and WPI.
Collapse
Affiliation(s)
- Mona A El-Banna
- Medical Biochemistry Department, National Research Centre, Dokki, Cairo, Egypt
| | - Omnia M Hendawy
- Clinical Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Aziza A El-Nekeety
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| | - Mosaad A Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt.
| |
Collapse
|
81
|
Zhang R, Liu T, Li W, Ma Z, Pei P, Zhang W, Yang K, Tao Y. Tumor microenvironment-responsive BSA nanocarriers for combined chemo/chemodynamic cancer therapy. J Nanobiotechnology 2022; 20:223. [PMID: 35549949 PMCID: PMC9097166 DOI: 10.1186/s12951-022-01442-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023] Open
Abstract
Tumor microenvironment (TME), characterized by high glutathione (GSH), high hydrogen peroxide (H2O2) and acidic pH levels, is favorable for the growth, invasion and metastasis of cancer cells. Taking advantage of the specific characteristics of tumors, TME-responsive GCBD NPs are designed to deliver nanoscale coordination polymers (NCPs, GA-Cu) and chemotherapy drugs (doxorubicin, DOX) based on bovine serum albumin (BSA) nanocarriers into cancer cells for combined chemodynamic therapy (CDT) and chemotherapy. In an acidic environment, GCBD NPs could release approximately 90% copper ions, which can not only consume overexpressed GSH to modulate the TME but can also react with endogenous H2O2 in a Fenton-like reaction to achieve the CDT effect. Meanwhile, the released DOX could enter the nucleus of tumor cells and affect their proliferation to achieve efficient chemotherapy. Both in vitro and in vivo experiments showed that GCBD NPs had good biosafety and could effectively inhibit the growth of cancer cells. GCBD NPs are promising as a biocompatible nanoplatform to exploit TME characteristics for combined chemo and chemodynamic therapy, providing a novel strategy to eradicate tumors with high efficiency and specificity.
Collapse
Affiliation(s)
- Ruiyi Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, China
| | - Teng Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Wanzhen Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, China
| | - Zhiyuan Ma
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, China
| | - Pei Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Weiwei Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, Anhui, China.
| |
Collapse
|
82
|
Huang CH, Zaenudin E, Tsai JJ, Kurubanjerdjit N, Ng KL. Network subgraph-based approach for analyzing and comparing molecular networks. PeerJ 2022; 10:e13137. [PMID: 35529499 PMCID: PMC9074881 DOI: 10.7717/peerj.13137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/28/2022] [Indexed: 01/12/2023] Open
Abstract
Molecular networks are built up from genetic elements that exhibit feedback interactions. Here, we studied the problem of measuring the similarity of directed networks by proposing a novel alignment-free approach: the network subgraph-based approach. Our approach does not make use of randomized networks to determine modular patterns embedded in a network, and this method differs from the network motif and graphlet methods. Network similarity was quantified by gauging the difference between the subgraph frequency distributions of two networks using Jensen-Shannon entropy. We applied the subgraph approach to study three types of molecular networks, i.e., cancer networks, signal transduction networks, and cellular process networks, which exhibit diverse molecular functions. We compared the performance of our subgraph detection algorithm with other algorithms, and the results were consistent, but other algorithms could not address the issue of subgraphs/motifs embedded within a subgraph/motif. To evaluate the effectiveness of the subgraph-based method, we applied the method along with the Jensen-Shannon entropy to classify six network models, and it achieves a 100% accuracy of classification. The proposed information-theoretic approach allows us to determine the structural similarity of two networks regardless of node identity and network size. We demonstrated the effectiveness of the subgraph approach to cluster molecular networks that exhibit similar regulatory interaction topologies. As an illustration, our method can identify (i) common subgraph-mediated signal transduction and/or cellular processes in AML and pancreatic cancer, and (ii) scaffold proteins in gastric cancer and hepatocellular carcinoma; thus, the results suggested that there are common regulation modules for cancer formation. We also found that the underlying substructures of the molecular networks are dominated by irreducible subgraphs; this feature is valid for the three classes of molecular networks we studied. The subgraph-based approach provides a systematic scenario for analyzing, compare and classifying molecular networks with diverse functionalities.
Collapse
Affiliation(s)
- Chien-Hung Huang
- Department of Computer Science and Information Engineering, National Formosa University, Yun-Lin, Taiwan
| | - Efendi Zaenudin
- National Research and Innovation Agency, Bandung, Jawa Barat, Republic of Indonesia,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Jeffrey J.P. Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | | | - Ka-Lok Ng
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan,Center for Artificial Intelligence and Precision Medicine Research, Asia University, Taichung, Taiwan,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
83
|
Zhang YY, Ren H, Yan QL, Li YL, Liu Q, Yao GD, Song SJ. SCP-7, a germacrane-type sesquiterpene lactone derivative, induces ROS-mediated apoptosis in NSCLC cells in vitro and in vivo. Eur J Pharmacol 2022; 925:174989. [PMID: 35490722 DOI: 10.1016/j.ejphar.2022.174989] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/16/2022]
Abstract
Scabertopin (SCP), an abundant germacrane-type sesquiterpene lactone (SLC) isolated from Elephantopus scaber, was selected as a reference compound for modification and evaluation as anticancer agents for non-small cell lung cancer (NSCLC) treatment. All derivatives (SCP-1-SCP-13) except for SCP-3 showed potential inhibitory effect (IC50 5.2-9.7 μM) against A549 cells. The most promising compound SCP-7 also showed good cytotoxic activity against another two NSCLC cell lines (H1299 and H460), with IC50 value of 4.4 and 8.9 μM, respectively. Furthermore, SCP-7 could induce apoptotic cell death that was associated with the increased reactive oxygen species (ROS) generation, the loss of mitochondrial membrane potential, Bcl-2 family proteins modulation, caspases-3 and PARP cleavage. In addition, SCP-7 also inhibited cell growth by increasing Bax expression and reducing the Ki-67 positive cells in vivo, but there were no obvious toxic and side effects on internal organs. Mechanistically, PharmMapper, molecular docking and Western blot analysis revealed that SCP-7 might interact with the epidermal growth factor receptor (EGFR) and inhibit its expression in lung cancer cells. Together, above results suggest further effective application of SCP-7 as a potential anti-tumor agent in the treatment of NSCLC.
Collapse
Affiliation(s)
- Yang-Yang Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Hui Ren
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Qiu-Lin Yan
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Ya-Ling Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|
84
|
Baicalein Inhibits the SMYD2/RPS7 Signaling Pathway to Inhibit the Occurrence and Metastasis of Lung Cancer. JOURNAL OF ONCOLOGY 2022; 2022:3796218. [PMID: 35432530 PMCID: PMC9012617 DOI: 10.1155/2022/3796218] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/23/2022] [Accepted: 03/03/2022] [Indexed: 12/23/2022]
Abstract
Objective This study investigated the potential effects of Baicalein on proliferation, migration, and invasion of human lung cancer A549 and NCI-H1299 cells and its possible mechanisms. Methods The effects of Baicalein on proliferation and invasion of A549 and NCI-H1299 cells were detected by MTT assay, clonogenesis assay, and Transwell assay. A subcutaneous transplanted tumor model was used to evaluate the effects of SMYD2 and Baicalein on the proliferation of lung cancer. Baicalein inhibited in SMYD2/RPS7 signaling pathway in tumor cells was also detected by qRT-PCR. Results Baicalein significantly inhibited the growth of lung cancer cells. In addition, Baicalein significantly reduced the rate of A549 and NCI-H1299 cell invasion and clone formation in a dose-dependent manner. Animal experiments showed that both SMYD2 and Baicalein treatments could inhibit lung cancer tumor proliferation. Mechanism studies suggest that Baicalein inhibits the SMYD2/RPS7 signaling pathway. Conclusion These results indicated that Baicalein could inhibit the proliferation, migration, and invasion of LUNG cancer A549 and NCI-H1299 cells. Baicalein inhibits cell proliferation by downregulating the SMYD2/RPS7 signaling pathway.
Collapse
|
85
|
Wang KN, Liu LY, Mao D, Hou MX, Tan CP, Mao ZW, Liu B. A Nuclear-Targeted AIE Photosensitizer for Enzyme Inhibition and Photosensitization in Cancer Cell Ablation. Angew Chem Int Ed Engl 2022; 61:e202114600. [PMID: 35132748 DOI: 10.1002/anie.202114600] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Indexed: 12/24/2022]
Abstract
The nucleus is considered the ideal target for anti-tumor therapy because DNA and some enzymes in the nucleus are the main causes of cell canceration and malignant proliferation. However, nuclear target drugs with good biosafety and high efficiency in cancer treatment are rare. Herein, a nuclear-targeted material MeTPAE with aggregation-induced emission (AIE) characteristics was developed based on a triphenylamine structure skeleton. MeTPAE can not only interact with histone deacetylases (HDACs) to inhibit cell proliferation but also damage telomere and nucleic acids precisely through photodynamic treatment (PDT). The cocktail strategy of MeTPAE caused obvious cell cycle arrest and showed excellent PDT anti-tumor activity, which offered new opportunities for the effective treatment of malignant tumors.
Collapse
Affiliation(s)
- Kang-Nan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, China.,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Liu-Yi Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, China
| | - Duo Mao
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Ming-Xuan Hou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| |
Collapse
|
86
|
Najafzadeh B, Motafakkerazad R, Najafi S, Amini M, Alemohammad H, Vasefifar P, Baradaran B. Nanog suppression enhanced the chemosensitivity of Human Non-Small-Cell Lung Cancer cells to Cisplatin and inhibited cell migration. Pathol Res Pract 2022; 233:153869. [DOI: 10.1016/j.prp.2022.153869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/25/2022]
|
87
|
Enhanced radiosensitivity by 6-thio-dG via increasing telomere dysfunction and ataxia telangiectasia mutated inhibition in non-small cell lung cancer. RADIATION MEDICINE AND PROTECTION 2022. [DOI: 10.1016/j.radmp.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
88
|
Mirzaeinia S, Zeinali S, Budisa N, Karbalaei-Heidari HR. Targeted Codelivery of Prodigiosin and Simvastatin Using Smart BioMOF: Functionalization by Recombinant Anti-VEGFR1 scFv. Front Bioeng Biotechnol 2022; 10:866275. [PMID: 35402395 PMCID: PMC8987009 DOI: 10.3389/fbioe.2022.866275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 11/18/2022] Open
Abstract
Biological metal-organic frameworks (BioMOFs) are hybrid compounds in which metal nodes are linked to biocompatible organic ligands and have potential for medical application. Herein, we developed a novel BioMOF modified with an anti-VEGFR1 scFv antibody (D16F7 scFv). Our BioMOF is co-loaded with a combination of an anticancer compound and a lipid-lowering drug to simultaneously suppress the proliferation, growth rate and metastases of cancer cells in cell culture model system. In particular, Prodigiosin (PG) and Simvastatin (SIM) were co-loaded into the newly synthesized Ca-Gly BioMOF nanoparticles coated with maltose and functionalized with a recombinant maltose binding protein-scFv fragment of anti-VEGFR1 (Ca-Gly-Maltose-D16F7). The nanoformulation, termed PG + SIM-NP-D16F7, has been shown to have strong active targeting behavior towards VEGFR1-overexpresing cancer cells. Moreover, the co-delivery of PG and SIM not only effectively inhibits the proliferation of cancer cells, but also prevents their invasion and metastasis. The PG + SIM-NP-D16F7 nanocarrier exhibited stronger cytotoxic and anti-metastatic effects compared to mono-treatment of free drugs and drug-loaded nanoparticles. Smart co-delivery of PG and SIM on BioMOF nanoparticles had synergistic effects on growth inhibition and prevented cancer cell metastasis. The present nanoplatform can be introduced as a promising tool for chemotherapy compared with mono-treatment and/or non-targeted formulations.
Collapse
Affiliation(s)
- Somayyeh Mirzaeinia
- Molecular Biotechnology Lab, Department of Biology, Faculty of Science, Shiraz University, Shiraz, Iran
| | - Sedighe Zeinali
- Department of Nanochemical Engineering, School of Advanced Technologies, Nanotechnology Research Institute, Shiraz University, Shiraz, Iran
| | - Nediljko Budisa
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB, Canada
- Institut für Chemie, Technische Universität Berlin, Berlin, Germany
| | - Hamid Reza Karbalaei-Heidari
- Molecular Biotechnology Lab, Department of Biology, Faculty of Science, Shiraz University, Shiraz, Iran
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
89
|
Hu P, Ni C, Teng P. Effects of artesunate on the malignant biological behaviors of non-small cell lung cancer in human and its mechanism. Bioengineered 2022; 13:6590-6599. [PMID: 35361045 PMCID: PMC9278965 DOI: 10.1080/21655979.2022.2042141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
We aimed to assess the effects of artesunate (ART) on the proliferation, migration, invasion and apoptosis of the non-small cell lung cancer cells A549 and H1299. The effects of ART and carboplatin (CBP) alone or in combination on the viability of A549 and H1299 cells were evaluated by MTT assay. The effects of 30 μg/ml ART on cell invasion, migration and apoptosis were evaluated by Transwell assay, scratch assay and flow cytometry, respectively. The protein expressions of human antigen R (HuR) and MMP-9 after treatment with 30 μg/ml ART for 48 h were detected by Western blotting. After 48 h of treatment, 9 μg/ml ART in combination with 7 μg/ml CBP exerted a mild synergistic effect on cell viability. The migration rates of cells treated with 30 μg/ml ART and number of invasive cells were significantly lower, and the apoptosis rates were higher than those of the DMSO-treated group. HuR and MMP-9 expressions in cells treated with 30 μg/ml ART for 48 h were significantly lower than those of the DMSO-treated group. ART suppresses the proliferation, migration and invasion of A549 and H1299 cells and induces their apoptosis, probably being associated with decreased expressions of HuR and MMP-9 proteins.
Collapse
Affiliation(s)
- Peng Hu
- Department of Cardiac and Great Vessel Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Chengyao Ni
- Department of Cardiac and Great Vessel Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Pen Teng
- Department of Cardiac and Great Vessel Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
90
|
Minchom A, Viteri S, Bazhenova L, Gadgeel SM, Ignatius Ou SH, Trigo J, Bauml JM, Backenroth D, Bhattacharya A, Li T, Mahadevia P, Girard N. Amivantamab compared with real-world therapies in patients with advanced non-small cell lung cancer harboring EGFR exon 20 insertion mutations who progressed after platinum-based chemotherapy. Lung Cancer 2022; 168:74-82. [DOI: 10.1016/j.lungcan.2022.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/19/2022]
|
91
|
Untargeted LC-MS/MS Metabolomics Study on the MCF-7 Cell Line in the Presence of Valproic Acid. Int J Mol Sci 2022; 23:ijms23052645. [PMID: 35269790 PMCID: PMC8910739 DOI: 10.3390/ijms23052645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
To target breast cancer (BC), epigenetic modulation could be a promising therapy strategy due to its role in the genesis, growth, and metastases of BC. Valproic acid (VPA) is a well-known histone deacetylase inhibitor (HDACi), which due to its epigenetic focus needs to be studied in depth to understand the effects it might elicit in BC cells. The aim of this work is to contribute to exploring the complete pharmacological mechanism of VPA in killing cancer cells using MCF-7. LC-MS/MS metabolomics studies were applied to MCF-7 treated with VPA. The results show that VPA promote cell death by altering metabolic pathways principally pentose phosphate pathway (PPP) and 2′deoxy-α-D-ribose-1-phosphate degradation related with metabolites that decrease cell proliferation and cell growth, interfere with energy sources and enhance reactive oxygen species (ROS) levels. We even suggest that mechanisms such as ferropoptosis could be involved due to deregulation of L-cysteine. These results suggest that VPA has different pharmacological mechanisms in killing cancer cells including apoptotic and nonapoptotic mechanisms, and due to the broad impact that HDACis have in cells, metabolomic approaches are a great source of information to generate new insights for this type of molecule.
Collapse
|
92
|
Wang K, Liu L, Mao D, Hou M, Tan C, Mao Z, Liu B. A Nuclear‐Targeted AIE Photosensitizer for Enzyme Inhibition and Photosensitization in Cancer Cell Ablation. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202114600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- Kang‐Nan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University China
- Department of Chemical and Biomolecular Engineering National University of Singapore 4 Engineering Drive 4 Singapore 117585 Singapore
| | - Liu‐Yi Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University China
| | - Duo Mao
- Department of Chemical and Biomolecular Engineering National University of Singapore 4 Engineering Drive 4 Singapore 117585 Singapore
| | - Ming‐Xuan Hou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University China
| | - Cai‐Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University China
| | - Zong‐Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry State Key Laboratory of Oncology in South China Sun Yat-Sen University China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering National University of Singapore 4 Engineering Drive 4 Singapore 117585 Singapore
| |
Collapse
|
93
|
Li J, Ma A, Lan W, Liu Q. Platycodon D-induced A549 cell apoptosis through RRM1-regulated p53/VEGF/MMP2 pathway. Anticancer Agents Med Chem 2022; 22:2458-2467. [PMID: 35088678 DOI: 10.2174/1871520622666220128095355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/05/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lung cancer is one of the leading causes of cancer-related deaths worldwide. Platycodin D (PD), a major pharmacological constituent from the Chinese medicinal herb named Platycodonis Radix, has shown potent anti-tumor activity. Also, it is also reported that PD could inhibit cellular growth in the non-small-cell lung carcinoma (NSCLC) A549 cell line. However, the underlying mechanism is not fully clarified. METHODS Cell proliferation was measured by MTT assay. Annexin V and propidium iodide (PI) assay were employed to study the apoptosis effects of PD on A549 cells. Western blot analysis was used to evaluate protein expression. Also, we used a siRNA against p53, as well as a plasmid-based RRM1 over-expression to investigate their functions. RESULTS It demonstrated PD inhibited A549 cell proliferation in a dose- and time-dependent manner. Further investigations showed that PD induced cell apoptosis, which was supported by dose-dependent and time-dependent caspase-3 activation and p53/VEGF/MMP2 pathway regulation. Also, PD demonstrated the inhibition effect of ribonucleotide reductase M1 (RRM1), whose role in various tumors is contradictory. Remarkably, in this work, RRM1 overexpression in A549 cells could have a negative impact on the regulation of the p53/VEGF/MMP2 pathway induced by PD treatment. Note as well that RRM1 overexpression also attenuated cell apoptosis and inhibition of cell proliferation of A549 treated with PD. CONCLUSION The results suggested that PD could inhibit A549 cell proliferation and induce cell apoptosis by regulating p53/VEGF/MMP2 pathway, in which RRM1 plays an important role directly.
Collapse
Affiliation(s)
- Jiurong Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P. R. of China
| | - Aiping Ma
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P. R. of China
| | - Wenbin Lan
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P. R. of China
| | - Qun Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P. R. of China
| |
Collapse
|
94
|
Swantara MD, Rita WS, Dira MA, Agustina KK. Cervical anticancer activities of Annona squamosa Linn. leaf isolate. Vet World 2022; 15:124-131. [PMID: 35369588 PMCID: PMC8924393 DOI: 10.14202/vetworld.2022.124-131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/20/2021] [Indexed: 11/17/2022] Open
Abstract
Background and Aim: Cancer is one of the leading causes of death, the need for new anticancer herbal drugs is becoming more urgent considering the side effects of synthetic drugs. This study aimed to determine the anticancer activity of isolates derived from the methanol extract of Annona squamosa Linn. leaves and to identify the compounds that have an active effect against HeLa cells. Materials and Methods: The leaf metabolites of A. squamosa L. were extracted using methanol at room temperature (28°C) and were partitioned into n-hexane, chloroform, and n-butanol. The toxicity test of these extracts was conducted using a brine shrimp lethality assay. Furthermore, the most toxic extracts were separated and purified using silica gel column chromatography to yield four isolate fractions: FA, FB, FC, and FD. The most toxic isolates were tested for anticancer against HeLa cells, and their compounds were identified using liquid chromatography-mass spectrometry. Results: The results showed that the most toxic isolate with an LC50 value of 100.00 ppm had a potency similar to that of an anticancer agent with an IC50 value of 70.9021 ppm. Furthermore, the five compounds identified in this isolate include (6S, 7aR)-6-hydroxy-4,4,7a-trimethyl-6,7-dihydro-5H-1-benzofuran-2-one or loliolide, cocamidopropyl betaine, N-[3-(dimethylamino)propyl]dodecanamide or lauramidopropyl dimethylamine, linolenic acid, and 1-dodecyl-2-azepanone or laurocapram. Conclusion: It can be concluded that the leaf isolates of A. squamosa Linn. had shown anticancer activities against cervical cancer.
Collapse
Affiliation(s)
- Made Dira Swantara
- Department of Applied Chemistry, Faculty of Mathematics and Natural Sciences, Udayana University, Denpasar, Bali 80225 Indonesia
| | - Wiwik Susanah Rita
- Department of Applied Chemistry, Faculty of Mathematics and Natural Sciences, Udayana University, Denpasar, Bali 80225 Indonesia
| | - Made Asmarani Dira
- Clinical and Community Pharmacy Study Program, Faculty of Health, Bali Institute of Technology and Health, Denpasar, Bali 80225, Indonesia
| | - Kadek Karang Agustina
- Department of Public Health, The Faculty of Veterinary Medicine, Udayana University, Denpasar, Bali 80225, Indonesia
| |
Collapse
|
95
|
Bhavya K, Mantipally M, Roy S, Arora L, Badavath VN, Gangireddy M, Dasgupta S, Gundla R, Pal D. Novel imidazo[1,2-a]pyridine derivatives induce apoptosis and cell cycle arrest in non-small cell lung cancer by activating NADPH oxidase mediated oxidative stress. Life Sci 2022; 294:120334. [PMID: 35065161 DOI: 10.1016/j.lfs.2022.120334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/29/2021] [Accepted: 01/14/2022] [Indexed: 12/30/2022]
Abstract
AIMS Imidazo[1,2-a]pyridine-based analogues have recently gained significant interest because of their wide spectrum of biological activities including anti-cancer potential, however the development of targeted therapeutic candidates against non-small cell lung cancer (NSCLC) is of utmost need due to its high prevalence and poor prognosis. Herein, we have aimed to synthesized novel imidazo [1,2-a] pyridine derivatives (IMPA) by coupling with 2-amino-4H-pyranto enhance bioactivity against NSCLC. MAIN METHODS We have designed and synthesized a series of fifteen novel imidazo [1,2-a] pyridine derivatives through molecular hybridization and studied their anti-cancer activity against in-vitro lung adenocarcinoma and 3D multicellular lung tumor spheroids. KEY FINDINGS IMPA-2, IMPA-5, IMPA-6, IMPA-8, and IMPA-12 markedly induced cytotoxicity by notably increased NADPH oxidase (NOX) activity, which results in the induction of ROS-mediated apoptosis in A549 lung cancer cells. It caused impairment of mitochondrial membrane potential by increasing pro-apoptotic BAX, and BAK1 expressions, and decreasing anti-apoptotic BCL2 expression, along with the induction of caspase-9/3 activation, however, these attributes were compromised in presence of N-acetyl-L-cysteine (NAC), a free radical scavenger. Increased ROS production by IMPAs also promotes p53 mediated cell cycle arrest through the inactivation of p38MAPK. Reduction of tumor size in IMPAs-treated 3D multicellular lung tumor spheroids gave further validation. SIGNIFICANCE Beside cytotoxicity, IMPAs also inhibit lung cancer cell invasion and migration, suggesting their applicability in metastatic lung cancer. Therefore, IMPA derivatives could be used as potential anti-cancer agents in treating non-small cell lung cancer.
Collapse
Affiliation(s)
- Kumari Bhavya
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Manohar Mantipally
- Department of Chemistry, School of Science, GITAM Deemed University, Hyderabad 502329, Telangana, India
| | - Soumyajit Roy
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Leena Arora
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Vishnu Nayak Badavath
- Institute for Drug Research, The Hebrew University, Jerusalem 9112001, Israel; Chitkara College of Pharmacy, Chitkara University, Punjab 140410, India
| | | | - Suman Dasgupta
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Sonitpur 784028, Assam, India
| | - Rambabu Gundla
- Department of Chemistry, School of Science, GITAM Deemed University, Hyderabad 502329, Telangana, India.
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India.
| |
Collapse
|
96
|
Malhotra S, Dumoga S, Mehta S, Rao EP, Mohanty S, Singh N. Engineering Cellular Membrane for Dual Mode Therapy Using NIR Responsive Photosensitizer and Reversible Topoisomerase Inhibition Activity. ACS APPLIED BIO MATERIALS 2022; 5:570-582. [PMID: 35040623 DOI: 10.1021/acsabm.1c01070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Extensive research over past few decades has highlighted the challenges of chemotherapy and prompted the need for multimodality therapy because chemotherapy alone cannot fully eradicate the tumor due to physiological barriers in its effective delivery and systemic side effects. It can be mitigated by adopting nanoparticles as more effective delivery method, but none of them completely prevents drug toxicities. Utilizing multiple therapeutic modes such as phototherapy that can act synergistically with chemotherapy in controlling tumor growth, while reducing the overall dosage, could become a preferred route for cancer management. Careful selection of nanoparticle system, which can simultaneously deliver both drug and photosensitizer, can significantly enhance the therapeutic outcome. Therefore, in this paper, we report development and potential of immune-compatible and long circulating nanoerythrosomes for enhancing the therapeutic potential of camptothecin and indocyanine green against murine cancer model. The RBCs membrane simultaneously loaded the nonpolar drug and amphiphilic photosensitizer in its lipid bilayer, which self-assembled to form stable nanoparticles. These nano constructs absorbed light in the near-infrared region and hence are suitable for targeting deep seated tissues. The dual chemo-phototherapy had great effect on cell viability and had therapeutic value.
Collapse
Affiliation(s)
- Sahil Malhotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Shweta Dumoga
- Meerut Institute of Engineering and Technology, Meerut 250005, India
| | - Supriya Mehta
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - E Pranshu Rao
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.,Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
97
|
Chinnaiyan SK, Pandiyan R, Natesan S, Chindam S, Gouti AK, Sugumaran A. Fabrication of basil oil Nanoemulsion loaded gellan gum hydrogel—evaluation of its antibacterial and anti-biofilm potential. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103129] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
98
|
Akinyelu J, Oladimeji O, Daniels A, Singh M. Folate-targeted doxorubicin delivery to breast and cervical cancer cells using a chitosan-gold nano-delivery system. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
99
|
Malik D, Mahendiratta S, Kaur H, Medhi B. Futuristic approach to cancer treatment. Gene 2021; 805:145906. [PMID: 34411650 DOI: 10.1016/j.gene.2021.145906] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/13/2021] [Indexed: 11/26/2022]
Abstract
Cancer is becoming one of the deadliest disease in both developed as well as developing countries and continuous effort is being made to find innovative therapies for myriad types of cancers that afflict the human body. Therapeutic options for cancer have grown exponentially over the time but we are quite a way off from finding a magic bullet that can help cure cancer and based on the current evidence we may never find a catch all cure ever and it becomes crucial that we keep on innovating and find multiple ways to attack the menace of this dreaded disease. Many patients suffer recurrence of disease and require second-line or in some cases more than two lines of treatment. In this review article we have discussed the available therapies along with the newer advancements that have been made in cancer therapy. Latest developments in treatment of various cancers that have been discussed include gene editing using CRISPR/Cas9, theranostics, viral mediated therapy, artificial intelligence, tumor infiltrating lymphocyte therapy, etc.
Collapse
Affiliation(s)
- Deepti Malik
- Department of Biochemistry, All India Institute of Medical Sciences, Bilaspur, India
| | - Saniya Mahendiratta
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harpinder Kaur
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
100
|
Hawash M, Jaradat N, Bawwab N, Salem K, Arafat H, Hajyousef Y, Shtayeh T, Sobuh S. Design, synthesis, and biological evaluation of phenyl-isoxazole-carboxamide derivatives as anticancer agents. HETEROCYCL COMMUN 2021. [DOI: 10.1515/hc-2020-0134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
The present study aimed to design and synthesize a series of phenyl-isoxazole-carboxamide derivatives and investigate their antitumor and antioxidant activities. The in vitro cytotoxic evaluation was conducted using the MTS assay against four cancer cell lines: hepatocellular carcinoma (Hep3B and HepG2), cervical adenocarcinoma (HeLa), breast carcinoma (MCF-7), in addition to the normal cell line (Hek293T). Besides, the antioxidant activity was evaluated using a 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay. All obtained compounds were found to have potent to moderate activities against Hep3B and MCF-7 cancer cells lines, except compound 2e. It was found that compound 2a has potent activity against HeLa and Hep3B cancer cell lines with IC50 values of 0.91 and 8.02 µM, respectively. The IC50 dose range of the tested compounds against Hep3B was 5.96–28.62 µM, except for 2e, compared with doxorubicin, which has an IC50 value of 2.23 µM. Also, the IC50 value range of the compounds against Hek293T was 112.78–266.66 µM, compared with doxorubicin, which has an IC50 dose of 0.581 µM. The antioxidant activity of the synthesized compounds was weak, and compound 2d showed moderate activity against the DPPH enzyme with an IC50 value of 138.50 µM in comparison with Trolox, which has an IC50 dose of 37.23 µM.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus P.O. Box 7, 00970 , Palestine
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus P.O. Box 7, 00970 , Palestine
| | - Noor Bawwab
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus P.O. Box 7, 00970 , Palestine
| | - Kamilah Salem
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus P.O. Box 7, 00970 , Palestine
| | - Hadeel Arafat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus P.O. Box 7, 00970 , Palestine
| | - Yousef Hajyousef
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus P.O. Box 7, 00970 , Palestine
- Department of Pharmacy, Faculty of Pharmacy, Cyprus International University , Lefkosa, TRNC, Via Mersin 10, 99258 , Turkey
| | - Tahrir Shtayeh
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus P.O. Box 7, 00970 , Palestine
| | - Shorooq Sobuh
- Department of Biomedical Sciences, Physiology, Pharmacology & Toxicology Division, Faculty of Medicine and Health Sciences, An-Najah National University , Nablus , Palestine
| |
Collapse
|