1
|
Wang M, Shi Z, Wang F, Wang C, Wang H. Integrating structure-activity relationships, computational approaches, and experimental validation to unlock the therapeutic potential of indole-3-carbinol and its derivatives. Biochem Pharmacol 2025; 238:116968. [PMID: 40318812 DOI: 10.1016/j.bcp.2025.116968] [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: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Indole-3-carbinol (I3C) a bioactive compound derived from cruciferous vegetables, has garnered significant attention for its role in cancer prevention and its broad-spectrum biological activities, including anti-inflammatory properties and the modulation of critical signaling pathways. This review explores the structure-activity relationship (SAR) of I3C and its derivatives, emphasizing their molecular mechanisms and therapeutic potential. Key cellular targets, such as estrogen receptors, and pathways, including NF-κB, Wnt/β-catenin, and PI3K/Akt, are highlighted for their roles in apoptosis, autophagy, and the disruption of mitogenic signaling. The SAR analysis reveals the influence of molecular modifications, particularly in dimeric forms like diindolylmethane (DIM) on pharmacokinetics and bioactivity. Computational approaches, including molecular docking, molecular dynamics simulations, and density functional theory (DFT) provide insights into ligand-receptor interactions, binding energetics, and electronic properties, facilitating biological activity predictions. Experimental evidence from in vitro assays and synergistic studies underscores the cytotoxic efficacy and combinatorial benefits of I3C with conventional chemotherapeutics. Challenges in clinical translation, such as bioavailability and targeted delivery, are addressed, highlighting the potential of integrating computational and experimental findings to develop novel I3C-based therapeutics. This comprehensive analysis positions I3C as a promising scaffold for designing effective agents against cancer and other diseases.
Collapse
Affiliation(s)
- Mingjie Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| | - Zihan Shi
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| | - Fangfang Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China
| | - Cunqin Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| | - Hongting Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| |
Collapse
|
2
|
Tian C, Deng S, Yang M, Bai B, Pan Y, Xie G, Zhao D, Wei L. Indole-3-carbinol and its main derivative 3,3'-diindolylmethane: Regulatory roles and therapeutic potential in liver diseases. Biomed Pharmacother 2024; 180:117525. [PMID: 39388997 DOI: 10.1016/j.biopha.2024.117525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Indole-3-carbinol (I3C), a compound found in cruciferous vegetables, has shown significant efficacy in treating both cancerous and non-cancerous diseases. Its primary derivative, 3,3'-diindolylmethane (DIM), formed during digestion, also exhibits similar therapeutic benefits. In liver disorders, I3C and DIM exhibit dual roles by inhibiting and promoting hepatocellular carcinoma (HCC) and providing relief for nonmalignant liver diseases, such as acute liver injury (ALI), hepatic fibrosis, nonalcoholic fatty liver disease (NAFLD), and alcohol-related liver disease (ALD). Mechanistically, I3C and DIM modulate various pathophysiological processes, including cell proliferation, apoptosis, oxidative stress, and lipogenesis. This review aims to enhance researchers' understanding of the regulatory roles of I3C and DIM in these liver diseases and explore the potential of plant-derived substances in liver disease treatment.
Collapse
Affiliation(s)
- Chao Tian
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Shizhou Deng
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Research and Development Department, Guangdong Longsee Biomedical Corporation, Guangzhou 510700, China
| | - Ming Yang
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Baochen Bai
- Department of Cardiology, Peking University People's hospital, Beijing 100044, China
| | - Yi Pan
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Gangqiao Xie
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongliang Zhao
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China.
| |
Collapse
|
3
|
Ramakrishna K, Krishnamurthy S. Indole-3-carbinol ameliorated the neurodevelopmental deficits in neonatal anoxic injury in rats. Int J Dev Neurosci 2023; 83:31-43. [PMID: 36259087 DOI: 10.1002/jdn.10234] [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: 01/11/2022] [Revised: 10/01/2022] [Accepted: 10/13/2022] [Indexed: 02/04/2023] Open
Abstract
Neonatal anoxia is linked to long-lasting neurodevelopmental deficits. Due to the lack of pharmacological intervention to treat neonatal anoxia, there is interest in finding new molecules for its treatment. Indole-3-carbinol (I3C) has shown neuroprotective effects in some disease conditions. However, the neuroprotective role of I3C in neonatal anoxia has not been explored. Consequently, we have investigated the effect of I3C on neonatal anoxia-induced brain injury and neurodevelopmental deficits. Rat pups after 30 h of birth were subjected to two episodes of anoxia (10 min in each) at a time interval of 24 h by flowing 100% nitrogen. I3C was administered within 30 min of the second episode of anoxia on a postnatal day (PND) 3 and continued for PND 9. Neurodevelopmental deficits, cortical mitochondrial membrane potential (MMP), opening of mitochondrial permeability transition pore (MPTP), electron transport chain (ETC) enzyme activities, oxidative stress, hypoxia-inducible factor-1α (HIF-1α) levels, histopathological changes, and apoptosis were measured. I3C treatment dose-dependently ameliorated the neurodevelopmental deficits and somatic growth in anoxic pups. I3C improved mitochondrial function by enhancing the MMP, mitochondrial ETC enzymes, and antioxidants. It blocked the MPTP opening and release of cytochrome C in anoxic pups. Further, I3C reduced the elevated cortical HIF-1α in neonatal anoxic pups. Furthermore, I3C ameliorated histopathological abnormalities and mitochondrial-mediated apoptotic indicators Cyt C, caspase-9, and caspase-3. Our study concludes that I3C improved neuronal development in anoxic pups by enhancing mitochondrial function, reducing HIF-1α, and mitigating apoptosis.
Collapse
Affiliation(s)
- Kakarla Ramakrishna
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, India.,Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, India
| |
Collapse
|
4
|
Abdollah MRA, Ali AA, Elgohary HH, Elmazar MM. Antiangiogenic drugs in combination with seaweed fucoidan: A mechanistic in vitro and in vivo study exploring the VEGF receptor and its downstream signaling molecules in hepatic cancer. Front Pharmacol 2023; 14:1108992. [PMID: 36874031 PMCID: PMC9982147 DOI: 10.3389/fphar.2023.1108992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers reported worldwide with poor morbidity and high mortality rates. HCC is a very vascular solid tumour as angiogenesis is not only a key driver for tumour progression but also an exciting therapeutic target. Our research investigated the use of fucoidan, a sulfated polysaccharide readily abundant in edible seaweeds commonly consumed in Asian diet due to their extensive health benefits. Fucoidan was reported to possess a strong anti-cancer activity, but its anti-angiogenic potential is still to be fully unraveled. Our research investigated fucoidan in combination with sorafenib (an anti-VEGFR tyrosine kinase inhibitor) and Avastin® (bevacizumab, an anti-VEGF monoclonal antibody) in HCC both in vitro and in vivo. In vitro on HUH-7 cells, fucoidan had a potent synergistic effect when combined with the anti-angiogenic drugs and significantly reduced HUH-7 cell viability in a dose dependent manner. Using the scratch wound assay to test cancer cell motility, sorafenib, A + F (Avastin and fucoidan) or S + F (sorafenib and fucoidan) treated cells consistently showed an unhealed wound and a significantly smaller %wound closure (50%-70%) versus untreated control (91%-100%) (p < 0.05, one-way ANOVA). Using RT-qPCR; fucoidan, sorafenib, A + F and S + F significantly reduced the expression of the pro-angiogenic PI3K/AKT/mTOR and KRAS/BRAF/MAPK pathways by up to 3 folds (p < 0.05, one-way ANOVA versus untreated control). While ELISA results revealed that in fucoidan, sorafenib, A + F and S + F treated cells, the protein levels of caspases 3, 8, and 9 was significantly increased especially in the S + F group showing 40- and 16-times higher caspase 3 and 8 protein levels, respectively (p < 0.05, one-way-ANOVA versus untreated control). Finally, in a DEN-HCC rat model, H&E staining revealed larger sections of apoptosis and necrosis in the tumour nodules of rats treated with the combination therapies and immunohistochemical analysis of the apoptotic marker caspase 3, the proliferation marker Ki67 and the marker for angiogenesis CD34 showed significant improvements when the combination therapies were used. Despite the promising findings reported herein that highlighted a promising chemomodulatory effect of fucoidan when combined with sorafenib and Avastin, further investigations are required to elucidate potential beneficial or adversary interactions between the tested agents.
Collapse
Affiliation(s)
- Maha R A Abdollah
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt.,Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
| | - Aya A Ali
- Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
| | - Hassnaa H Elgohary
- Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
| | - Mohamed M Elmazar
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt.,Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
| |
Collapse
|
5
|
Indole-based hydrazone derivatives: Synthesis, cytotoxicity assessment, and molecular modeling studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133936] [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]
|
6
|
Machado ART, Tuttis K, Santos PWDS, Aissa AF, Antunes LMG. Diallyl Disulfide Induces Chemosensitization to Sorafenib, Autophagy, and Cell Cycle Arrest and Inhibits Invasion in Hepatocellular Carcinoma. Pharmaceutics 2022; 14:pharmaceutics14122582. [PMID: 36559076 PMCID: PMC9788602 DOI: 10.3390/pharmaceutics14122582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Hepatocellular carcinoma is the seventh most common type of cancer in the world, with limited treatment options. A promising strategy to treat cancer is to associate chemotherapeutics and plant bioactive compounds. Here, we examined whether diallyl disulfide (DADS; 50-200 μM) and sorafenib (SORA; 8 μM), either alone or in combination, were toxic to hepatocellular carcinoma cells (HepG2) in vitro. We assessed whether DADS and/or SORA induced cell death (LIVE/DEAD assay and autophagy) and cell cycle changes (flow cytometry), altered expression of key genes and proteins (RT-qPCR and Western blot), and modulated tumorigenesis signatures, such as proliferation (clonogenic assay), migration (wound healing), and invasion (inserts). The DADS + SORA combination elicited autophagic cell death by upregulating LC3 and NRF2 expression and downregulating FOS and TNF expression; induced the accumulation of cells in the G1 phase which thereby upregulated the CHEK2 expression; and inhibited invasion by downregulating the MMP2 expression. Predictive analysis indicated the participation of the MAPK pathway in the reported results. The DADS + SORA combination suppressed both cell invasion and clonogenic survival, which indicated that it dampened tumor growth, proliferation, invasion, and metastatic potential. Therefore, the DADS + SORA combination is a promising therapy to develop new clinical protocols.
Collapse
Affiliation(s)
- Ana Rita Thomazela Machado
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Katiuska Tuttis
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Patrick Wellington da Silva Santos
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Alexandre Ferro Aissa
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
- Correspondence: ; Tel.: +55-16-3315-4725
| |
Collapse
|
7
|
Taha AM, Aboulwafa MM, Zedan H, Helmy OM. Ramucirumab combination with sorafenib enhances the inhibitory effect of sorafenib on HepG2 cancer cells. Sci Rep 2022; 12:17889. [PMID: 36284117 PMCID: PMC9596484 DOI: 10.1038/s41598-022-21582-w] [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: 04/15/2022] [Accepted: 09/29/2022] [Indexed: 01/20/2023] Open
Abstract
Sorafenib, an oral multiple kinase inhibitor, is the standardized treatment for hepatocellular carcinoma (HCC). One strategy to improve HCC therapy is to combine agents that target key signaling pathways. In this study we set out to investigate the effect of combining sorafenib with either bevacizumab (anti-VEGF), panitumumab (anti-EGFR) or ramucirumab (anti-VEGFR2) on HepG2 cancer cell line with the aim of improving efficacy and possibility of therapeutic dose reduction of sorafenib.: HepG2 cancer cell line was treated with sorafenib alone or in combination with either bevacizumab, panitumumab or ramucirumab. Cell proliferation; apoptosis and cell cycle distribution; gene expression of VEGFR2, EGFR, MMP-9 and CASPASE3; the protein levels of pVEGFR2 and pSTAT3 and the protein expression of CASPASE3, EGFR and VEGFR2 were determined. Combined treatments of sorafenib with ramucirumab or panitumumab resulted in a significant decrease in sorafenib IC50. Sorafenib combination with ramucirumab or bevacizumab resulted in a significant arrest in pre-G and G0/G1 cell cycle phases, significantly induced apoptosis and increased the relative expression of CASPASE3 and decreased the anti-proliferative and angiogenesis markers´ MMP-9 and pVEGFR2 or VEGFR2 in HepG2 cells. A significant decrease in the levels of pSTAT3 was only detected in case of sorafenib-ramucirumab combination. The combined treatment of sorafenib with panitumumab induced a significant arrest in pre-G and G2/M cell cycle phases and significantly decreased the relative expression of EGFR and MMP-9. Sorafenib-ramucirumab combination showed enhanced apoptosis, inhibited proliferation and angiogenesis in HepG2 cancer cells. Our findings suggest that ramucirumab can be a useful as an adjunct therapy for improvement of sorafenib efficacy in suppression of HCC.
Collapse
Affiliation(s)
| | - Mohammad Mabrouk Aboulwafa
- grid.7269.a0000 0004 0621 1570Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Al Khalifa Al Ma’moun St., Abbassia, Cairo, Egypt ,Present Address: Faculty of Pharmacy, King Salman International University, Ras-Sudr, South Sinai Egypt
| | - Hamdallah Zedan
- grid.7776.10000 0004 0639 9286Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Kasr El-eini St., Cairo, Egypt
| | - Omneya Mohamed Helmy
- grid.7776.10000 0004 0639 9286Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Kasr El-eini St., Cairo, Egypt
| |
Collapse
|
8
|
Cakil YD, Ozunal ZG, Kayali DG, Aktas RG, Saglam E. Anti-proliferative effects of paroxetine alone or in combination with sorafenib in HepG2 cells. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e201148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
9
|
Nurcahyanti ADR, Jap A, Lady J, Prismawan D, Sharopov F, Daoud R, Wink M, Sobeh M. Function of selected natural antidiabetic compounds with potential against cancer via modulation of the PI3K/AKT/mTOR cascade. Biomed Pharmacother 2021; 144:112138. [PMID: 34750026 DOI: 10.1016/j.biopha.2021.112138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/19/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with growing global incidence, as 387 million people were diagnosed in 2014 with an expected projection of 642 million in 2040. Several complications are associated with DM including heart attack, stroke, kidney failure, blindness, and cancer. The latter is the second leading cause of death worldwide accounting for one in every six deaths, with liver, pancreas, and endometrium cancers are the most abundant among patients with diabetes. Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway plays a vital role in developing a wide array of pathological disorders, among them diabetes and cancer. Natural secondary metabolites that counteract the deleterious effects of reactive oxygen species (ROS) and modulate PI3K/Akt/mTOR pathway could be a promising approach in cancer therapy. Here, 717 medicinal plants with antidiabetic activities were highlighted along with 357 bioactive compounds responsible for the antidiabetic activity. Also, 43 individual plant compounds with potential antidiabetic activities against cancer via the modulation of PI3K/Akt/mTOR cascade were identified. Taken together, the available data give an insight of the potential of repurposing medicinal plants and/or the individual secondary metabolites with antidiabetic activities for cancer therapy.
Collapse
Affiliation(s)
- Agustina Dwi Retno Nurcahyanti
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia.
| | - Adeline Jap
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Jullietta Lady
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Deka Prismawan
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Farukh Sharopov
- Chinese-Tajik Innovation Center for Natural Products, National Academy of Sciences of Tajikistan, Ayni str. 299/2, 734063, Dushanbe, Tajikistan
| | - Rachid Daoud
- African Genome Center, Mohammed VI Polytechnic University (UM6P), Lot 660, Hay Moulay Rachid, Ben Guerir 43150, Morocco
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Mansour Sobeh
- AgroBiosciences Research, Mohammed VI Polytechnic University, Lot 660-Hay Moulay Rachid, 43150 Ben-Guerir, Morocco.
| |
Collapse
|
10
|
Dahiya M, Dureja H. Sorafenib for hepatocellular carcinoma: potential molecular targets and resistance mechanisms. J Chemother 2021; 34:286-301. [PMID: 34291704 DOI: 10.1080/1120009x.2021.1955202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most widespread typical therapy-resistant, unresectable type of malignant solid tumour with a high death rate constituting huge medical concern. Sorafenib is a small molecule oral multi-target kinase potent inhibitor that acts by suppressing/blocking the multiplication of the tumour cells, angiogenesis, and encouraging apoptosis of the tumour cells. Though, the precise mechanism of tumour cell death induction by sorafenib is yet under exploration. Furthermore, genetic heterogeneity plays a critical role in developing sorafenib resistance, which leads the way to identify the need for predictive biomarkers responsible for drug resistance. Therefore, it is essential to find out the fundamental resistance mechanisms to expand therapeutic plans. The authors summarize the molecular concepts of resistance, progression, potential molecular targets, HCC management therapies, and discussion on the advancements expected in the coming future, inclusive of biomarker-driven treatment strategies, which may provide the prospects to design innovative therapeutically targeted strategies for the HCC treatment and the clinical implementation of emerging targeted agents.
Collapse
Affiliation(s)
- Mandeep Dahiya
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
11
|
Khatoon E, Banik K, Harsha C, Sailo BL, Thakur KK, Khwairakpam AD, Vikkurthi R, Devi TB, Gupta SC, Kunnumakkara AB. Phytochemicals in cancer cell chemosensitization: Current knowledge and future perspectives. Semin Cancer Biol 2020; 80:306-339. [DOI: 10.1016/j.semcancer.2020.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
|
12
|
López-García G, Cilla A, Barberá R, Alegría A. Anti-Inflammatory and Cytoprotective Effect of Plant Sterol and Galactooligosaccharides-Enriched Beverages in Caco-2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1862-1870. [PMID: 31290324 DOI: 10.1021/acs.jafc.9b03025] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Plant sterol (PS) (1 g/100 mL) enriched milk-based fruit beverages with or without galactooligosaccharides (GOS) (1.8 g/100 mL) were used in differentiated Caco-2 cells. Their potential cytopreventive effect against oxidative stress induced by cholesterol oxidation products (COPs) and their anti-inflammatory properties were evaluated. Preincubation (24 h) with bioaccessible fractions from beverages without and with GOS (MfB and MfB-G) completely prevented the COPs (60 μM/4 h) induced oxidative stress independent to GOS presence with exception to calcium influx and GSH content, where a partial protective effect was observed. Besides, MfB produced a significant (p < 0.05) reduction of IL-8 (40%) and IL-6 (50%) after IL-1β-induction (1 ng/mL/24 h) through the inhibition of NF-κB p65 translocation into the nucleus (16%) compared to control cells, while GOS presence compromised their anti-inflammatory effect. Therefore, PS-enriched milk-based fruit beverage could be an interesting strategy to prevent intestinal injury produced by COPs and to attenuate the pro-inflammatory process in intestinal human diseases. GOS addition had no extra beneficial antioxidant effect and even reduced their anti-inflammatory properties.
Collapse
Affiliation(s)
- Gabriel López-García
- Nutrition and Food Science Area, Faculty of Pharmacy , University of Valencia , Avenida Vicente Andrés Estellés/n , Burjassot, Valencia 46100 , Spain
| | - Antonio Cilla
- Nutrition and Food Science Area, Faculty of Pharmacy , University of Valencia , Avenida Vicente Andrés Estellés/n , Burjassot, Valencia 46100 , Spain
| | - Reyes Barberá
- Nutrition and Food Science Area, Faculty of Pharmacy , University of Valencia , Avenida Vicente Andrés Estellés/n , Burjassot, Valencia 46100 , Spain
| | - Amparo Alegría
- Nutrition and Food Science Area, Faculty of Pharmacy , University of Valencia , Avenida Vicente Andrés Estellés/n , Burjassot, Valencia 46100 , Spain
| |
Collapse
|
13
|
Salehi B, Zucca P, Sharifi-Rad M, Pezzani R, Rajabi S, Setzer WN, Varoni EM, Iriti M, Kobarfard F, Sharifi-Rad J. Phytotherapeutics in cancer invasion and metastasis. Phytother Res 2018; 32:1425-1449. [DOI: 10.1002/ptr.6087] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/11/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Bahare Salehi
- Medical Ethics and Law Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Paolo Zucca
- Department of Biomedical Sciences; University of Cagliari; Cagliari Italy
| | - Mehdi Sharifi-Rad
- Department of Medical Parasitology; Zabol University of Medical Sciences; Zabol 61663-335 Iran
| | - Raffaele Pezzani
- OU Endocrinology, Dept. Medicine (DIMED); University of Padova; via Ospedale 105 Padova 35128 Italy
- AIROB, Associazione Italiana per la Ricerca Oncologica di Base; Padova Italy
| | - Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - William N. Setzer
- Department of Chemistry; University of Alabama in Huntsville; Huntsville AL 35899 USA
| | - Elena Maria Varoni
- Department of Biomedical, Surgical and Dental Sciences; Milan State University; Milan Italy
| | - Marcello Iriti
- Department of Agricultural and Environmental Sciences; Milan State University; Milan Italy
| | - Farzad Kobarfard
- Phytochemistry Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Medicinal Chemistry, School of Pharmacy; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Javad Sharifi-Rad
- Phytochemistry Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Chemistry, Richardson College for the Environmental Science Complex; The University of Winnipeg; Winnipeg MB Canada
| |
Collapse
|
14
|
Min KJ, Um HJ, Kim JI, Kwon TK. The coffee diterpene kahweol enhances sensitivity to sorafenib in human renal carcinoma Caki cells through down-regulation of Mcl-1 and c-FLIP expression. Oncotarget 2017; 8:83195-83206. [PMID: 29137334 PMCID: PMC5669960 DOI: 10.18632/oncotarget.20541] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/30/2017] [Indexed: 01/07/2023] Open
Abstract
Sorafenib is approved for the treatment of hepatocellular carcinoma (HCC) and advanced renal cell carcinoma (RCC). However, low tumor response and side effects have been widely reported. Therefore, to improve the efficacy of sorafenib, we investigated whether combined treatment with sorafenib and kahweol, the coffee-specific diterpene, has a synergistic effect on apoptotic cell death. Combined treatment with sorafenib and kahweol markedly induced caspase-mediated apoptosis in renal carcinoma Caki cells. Combined treatment with sorafenib and kahweol induced down-regulation of Mcl-1 and c-FLIP expression. We found down-regulation of Mcl-1 and c-FLIP expression was modulated by the ubiquitin-proteasome pathway. Ectopic expression of Mcl-1 inhibited sorafenib plus kahweol-induced apoptosis. Interestingly, combined treatment with sorafenib and kahweol induced apoptotic cell death in c-FLIP overexpressed cells. In addition, combined treatment with sorafenib and kahweol markedly induced apoptosis in human lung carcinoma (A549) and breast carcinoma (MDA-MB-361) cells, but not in human normal mesangial cells and human skin fibroblast cells (HSF). Collectively, our study demonstrates that combined treatment with sorafenib and kahweol induces apoptotic cell death through down-regulation of Mcl-1 expression.
Collapse
Affiliation(s)
- Kyoung-Jin Min
- Department of Immunology, School of Medicine, Keimyung University, Daegu 704-701, South Korea
| | - Hee Jung Um
- Department of Immunology, School of Medicine, Keimyung University, Daegu 704-701, South Korea
| | - Jee In Kim
- Department of Molecular Medicine, School of Medicine, Keimyung University, Daegu 704-701, South Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 704-701, South Korea
| |
Collapse
|
15
|
Ziogas IA, Tsoulfas G. Evolving role of Sorafenib in the management of hepatocellular carcinoma. World J Clin Oncol 2017; 8:203-213. [PMID: 28638790 PMCID: PMC5465010 DOI: 10.5306/wjco.v8.i3.203] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/03/2017] [Accepted: 04/23/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant diseases worldwide and comes third in cancer-related mortality. Although there is a broad spectrum of treatment options to choose from, only a few patients are eligible candidates to receive a curative therapy according to their stage of disease, and thus palliative treatment is implemented in the majority of the patients suffering from liver cancer. Sorafenib, a multikinase inhibitor, is the only currently approved agent for systemic therapy in patients with advanced stage HCC and early stage liver disease. It has been shown to improve the overall survival, but with various side effects, while its cost is not negligible. Sorafenib has been in the market for a decade and has set the stage for personalized targeted therapy. Its role during this time has ranged from monotherapy to neoadjuvant and adjuvant treatment with surgical resection, liver transplantation and chemoembolization or even in combination with other chemotherapeutic agents. In this review our aim is to highlight in depth the current position of Sorafenib in the armamentarium against HCC and how that has evolved over time in its use either as a single agent or in combination with other therapies.
Collapse
|