1
|
Mohammadi F, Nejatollahi M, Sheikhnia F, Ebrahimi Y, Mohammadi M, Rashidi V, Alizadeh-Fanalou S, Azizzadeh B, Majidinia M. MiRNAs: main players of cancer drug resistance target ABC transporters. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6239-6291. [PMID: 39808313 DOI: 10.1007/s00210-024-03719-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/08/2024] [Indexed: 01/16/2025]
Abstract
Chemotherapy remains the cornerstone of cancer treatment; however, its efficacy is frequently compromised by the development of chemoresistance. Multidrug resistance (MDR), characterized by the refractoriness of cancer cells to a wide array of chemotherapeutic agents, presents a significant barrier to achieving successful and sustained cancer remission. One critical factor contributing to this chemoresistance is the overexpression of ATP-binding cassette (ABC) transporters. Furthermore, additional mechanisms, such as the malfunctioning of apoptosis, alterations in DNA repair systems, and resistance mechanisms inherent to cancer stem cells, exacerbate the issue. Intriguingly, microRNAs (miRNAs) have demonstrated potential in modulating chemoresistance by specifically targeting ABC transporters, thereby offering promising new avenues for overcoming drug resistance. This narrative review aims to elucidate the molecular underpinnings of drug resistance, with a particular focus on the roles of ABC transporters and the regulatory influence of miRNAs on these transporters.
Collapse
Affiliation(s)
- Forogh Mohammadi
- Department of Veterinary, Agriculture Faculty, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Masoumeh Nejatollahi
- Research Center for High School Students, Education System Zanjan Province, Zanjan, Iran
| | - Farhad Sheikhnia
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yaser Ebrahimi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Rashidi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahin Alizadeh-Fanalou
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Bita Azizzadeh
- Department of Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
2
|
Ashique S, Kumar P, Taj T, Debnath B, Mukherjee S, Patel A, Sridhar SB, Panigrahy UP, Poonia P, Selim S, Hussain MS. Nanotechnology: A State of the Art for the Management of Ocular Disorders—A Roadmap. BIONANOSCIENCE 2025; 15:285. [DOI: 10.1007/s12668-025-01895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2025] [Indexed: 05/04/2025]
|
3
|
Manjili DA, Babaei FN, Younesirad T, Ghadir S, Askari H, Daraei A. Dysregulated circular RNA and long non-coding RNA-Mediated regulatory competing endogenous RNA networks (ceRNETs) in ovarian and cervical cancers: A non-coding RNA-Mediated mechanism of chemotherapeutic resistance with new emerging clinical capacities. Arch Biochem Biophys 2025; 768:110389. [PMID: 40090441 DOI: 10.1016/j.abb.2025.110389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Cervical cancer (CC) and ovarian cancer (OC) are among the most common gynecological cancers with significant mortality in women, and their incidence is increasing. In addition to the prominent role of the malignant aspect of these cancers in cancer-related women deaths, chemotherapy drug resistance is a major factor that contributes to their mortality and presents a clinical obstacle. Although the exact mechanisms behind the chemoresistance in these cancers has not been revealed, accumulating evidence points to the dysregulation of non-coding RNAs (ncRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as key contributors. These ncRNAs perform the roles of regulators of signaling pathways linked to tumor formation and chemoresistance. Strong data from various recent studies have uncovered that the main mechanism of these ncRNAs in the induction of chemoresistance of CC and OC is done through a dysregulated miRNA sponge activity as competing endogenous RNA (ceRNA) in the competing endogenous RNA networks (ceRNETs), where a miRNA regulating a messenger RNA (mRNA) is trapped, thereby removing its inhibitory effect on the desired mRNA. Understanding these mechanisms is essential to enhancing treatment outcomes and managing the problem of drug resistance. This review provides a comprehensive overview of lncRNA- and circRNA-mediated ceRNETs as the core process of chemoresistance against the commonly used chemotherapeutics, including cisplatin, paclitaxel, oxaliplatin, carboplatin, and docetaxel in CC and OC. Furthermore, we highlight the clinical potential of these ncRNAs serving as diagnostic indicators of chemotherapy responses and therapeutic targets.
Collapse
Affiliation(s)
- Danial Amiri Manjili
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Naghdi Babaei
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tayebeh Younesirad
- Department of Medical Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sara Ghadir
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Askari
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
4
|
Mosoane B, McCabe M, Jackson BS, Dlamini Z. CD44 Variant Expression in Follicular Cell-Derived Thyroid Cancers: Implications for Overcoming Multidrug Resistance. Molecules 2025; 30:1899. [PMID: 40363706 PMCID: PMC12073131 DOI: 10.3390/molecules30091899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Thyroid cancer (TC) is a significant global health issue that exhibits notable heterogeneity in incidence and outcomes. In low-resource settings such as Africa, delayed diagnosis and limited healthcare access exacerbate mortality rates. Among follicular cell-derived thyroid cancers-including papillary (PTC), follicular (FTC), anaplastic (ATC), and poorly differentiated (PDTC) subtypes-the role of CD44 variants has emerged as a critical factor influencing tumor progression and multidrug resistance (MDR). CD44, a transmembrane glycoprotein, and its splice variants (CD44v) mediate cell adhesion, migration, and survival, contributing to cancer stem cell (CSC) maintenance and therapy resistance. Differential expression patterns of CD44 isoforms across TC subtypes have shown diagnostic, prognostic, and therapeutic implications. Specifically, CD44v6 expression in PTC has been correlated with metastasis and aggressive tumor behavior, while in FTC, its expression aids in distinguishing malignant from benign lesions. Furthermore, CD44 contributes to MDR through enhanced drug efflux via ABC transporters, apoptosis evasion, and CSC maintenance via the Wnt/β-catenin and PI3K/Akt pathways. Targeted therapies against CD44 such as monoclonal antibodies, hyaluronic acid-based nanocarriers, and gene-editing technologies hold promise in overcoming MDR. However, despite the mounting evidence supporting CD44-targeted strategies in various cancers, research on this therapeutic potential in TC remains limited. This review synthesizes existing knowledge on CD44 variant expression in follicular cell-derived thyroid cancers and highlights potential therapeutic strategies to mitigate MDR, particularly in high-burden regions, thereby improving patient outcomes and survival.
Collapse
Affiliation(s)
- Benny Mosoane
- Department of Anatomical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Michelle McCabe
- Department of Anatomical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Brandon S. Jackson
- Breast and Endocrine Unit, Department of General Surgery, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa;
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0001, South Africa
| |
Collapse
|
5
|
Asante JJ, Barger SW. P-glycoprotein and Alzheimer's Disease: Threats and Opportunities. ASN Neuro 2025; 17:2495632. [PMID: 40264334 DOI: 10.1080/17590914.2025.2495632] [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: 09/06/2024] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects more than 50 million people worldwide. One of the hallmark features of AD is the accumulation of amyloid β-peptide (Aβ) protein in the brain. P-glycoprotein (P-gp) is a membrane-bound protein expressed in various tissues, including the cerebrovascular endothelium. It plays a crucial role in the efflux of toxic substances, including Aβ, from the brain. Aberrations in P-gp levels or activity have been implicated in the pathogenesis of AD by promoting the accumulation of Aβ in the brain. Therefore, modulating the P-gp function represents a promising therapeutic strategy for treating AD. P-gp has multiple substrate binding sites, creating the potential for substrates to fall into complementation groups based on these sites; two substrates in the same complementation group may compete with one other, but two substrates in different groups may exhibit cooperativity. Thus, a given P-gp substrate may interfere with Aβ efflux whereas another may promote clearance. These threats and opportunities, as well as other aspects of P-gp relevance to AD, are discussed here.
Collapse
Affiliation(s)
- Joseph Jr Asante
- Graduate Program in Bioinformatics, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Neuroscience, Little Rock, AR, USA
- Geriatric Research, Education & Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| |
Collapse
|
6
|
Wang Y, Ding N, Zhao Y, Wang F, Liu W, Chen Z, Sun W, Gu L, Zhang Y. Design, synthesis, and biological evaluation of β-carboline derivatives as ABCB1 inhibitors for reversing multidrug resistance. Eur J Med Chem 2025; 288:117390. [PMID: 39965407 DOI: 10.1016/j.ejmech.2025.117390] [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: 12/20/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
The scarcity of ATP-binding cassette subfamily B member 1 (ABCB1, also known as P-glycoprotein, P-gp) inhibitors suitable for clinical application in improving multidrug resistance (MDR) promotes the development of drugs aimed at reversing MDR. In this work, we reported a comprehensive study for the first time about the reversal activity of β-carboline derivatives on ABCB1-mediated MDR. Among 48 synthesized derivatives, compound K27 significantly increased the sensitivity of ABCB1-mediated MDR SW620/AD300 cells to paclitaxel (PTX) (IC50 = 15.33 ± 5.4 nM, RF = 171.2) and hardly showed toxicity even at a high concentration of 20 μM when used alone. The in vitro studies indicated that compound K27 distinctly enhanced the arresting effect of PTX on the SW620/AD300 cell cycle, thereby inhibiting their proliferation. Mechanistically, compound K27 was confirmed to directly bind to ABCB1 to inhibit efflux function, reducing cellular efflux and ensuring stable intracellular concentration of PTX without affecting ABCB1's normal expression. Importantly, the combination of compound K27 and PTX exhibited potent tumor suppression in vivo without generating toxicity. These results demonstrated that β-carboline compounds represented by compound K27 may be potent ABCB1 inhibitors with considerable potential in effectively reversing ABCB1-mediated MDR, showing promising prospects.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Nanjin Ding
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yunpeng Zhao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengqing Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhe Chen
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiguang Sun
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Lianghu Gu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
7
|
Chang YT, Wu IT, Lee CH, Hung CC. Phyto-Sesquiterpene Lactones Prevent the Development of Multidrug Resistance in TNBC via ABC Transporters Inhibition and STAT3/MYC Signaling. Cancers (Basel) 2025; 17:1321. [PMID: 40282497 PMCID: PMC12026016 DOI: 10.3390/cancers17081321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Multidrug resistance (MDR) in triple-negative breast cancer (TNBC) leads to treatment failure and tumor recurrence. Dysregulation of the MYC oncogene is associated with the pathogenesis of TNBC and the development of chemoresistance via overexpression of ATP-binding cassette (ABC) transporters. Therefore, in the present study, we aimed to identify molecules from a natural product origin that prevent the development of MDR in TNBC by targeting the MYC signaling. METHODS The cell viability of TNBC was evaluated using sulforhodamine assay. Protein levels were detected by western blots or enzyme-linked immunosorbent assays. Intracellular calcein and hoechst33342 accumulation assay aimed to evaluate the inhibitory ability of phytocompounds on drug-efflux functions of ABCB1 and ABCG2 transporters. The Cancer Genome Atlas (TCGA) database was used to explore clinical genomic data. Furthermore, the zebrafish xenotransplantation model bearing Dil-labeled TNBC cells was applied to testify the in vivo effects of phyto-sesquiterpene lactones. RESULTS The results of the present study demonstrated that the phyto-sesquiterpene lactones exhibited an MDR prevention effect by repressing efflux activities of ABCB1 and ABCG2 transporters. Mechanistic studies showed that phyto-sesquiterpene lactones inducted TNBC cell apoptosis and cell cycle G2/M arrested by blocking the STAT3/MYC pathway. Clinical genomic data demonstrated that the percentages of MYC amplification and mRNA were upregulated approximately two-fold higher in the TNBC patients than the non-TNBC breast cancer patients. The survival of patients with an alteration in MYC was significantly lower in TNBC as compared to other subtypes. Moreover, the results of the zebrafish xenograft model confirmed that phyto-sesquiterpene lactones exerted stronger inhibitory effects on TNBC tumor growth in vivo. CONCLUSIONS In conclusion, these three phyto-sesquiterpene lactones were promising candidates for TNBC treatment and shed light on the prevention of developing MDR TNBC.
Collapse
Affiliation(s)
- Ying-Tzu Chang
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan; (Y.-T.C.); (I.-T.W.)
| | - I-Ting Wu
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan; (Y.-T.C.); (I.-T.W.)
| | - Chien-Hsing Lee
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chin-Chuan Hung
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan; (Y.-T.C.); (I.-T.W.)
- Department of Pharmacy, China Medical University Hospital, Taichung 404327, Taiwan
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
8
|
Khunweeraphong N, Kuchler K. The human ABCG2 transporter engages three gates to control multidrug extrusion. iScience 2025; 28:112125. [PMID: 40165990 PMCID: PMC11957596 DOI: 10.1016/j.isci.2025.112125] [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: 07/29/2024] [Revised: 01/17/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
The human ABCG2 transporter plays roles in physiological detoxification across barriers and in anticancer multidrug resistance. The translocation pathway for drug extrusion and its gating mechanism remains elusive. Here, we demonstrate that the ABCG2 multidrug transporter holds two cavities that are delineated by three regulatory gates, indicating a substrate translocation channel. Drugs are trapped in the central cavity after entering through the pivotal intracellular entry gate. This flexible cavity is surrounded by a cluster of three highly conserved phenylalanines. Their aromatic side chains enact a "clamp-push-seal" motion to ensure unidirectional substrate movement. The unique residues T435 and N436 act as critical selectors for ligands, determining the broad substrate specificity. The upper cavity is covered by the lid architecture, constituting the final gate before multidrug extrusion. This work unravels deep mechanistic details on how the translocation channel utilizes pivotal gating steps, including the sequence of events that drive ABCG2-mediated multidrug efflux.
Collapse
Affiliation(s)
- Narakorn Khunweeraphong
- Medical University of Vienna, Max Perutz Labs Vienna, Center for Medical Biochemistry Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Max Perutz Labs Vienna, Center for Medical Biochemistry Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| |
Collapse
|
9
|
do Nascimento Martinez L, da Silva MA, Fialho SN, Almeida ML, Dos Santos Ferreira A, de Jesus Gouveia A, do Nascimento WDSP, Dos Santos APDA, Rossi NRDLP, de Medeiros JF, Araújo NF, de Santana QLO, Kaiser CR, Ferreira SB, da Silva Araujo M, Teles CBG. In vitro and in silico evaluation of synthetic compounds derived from bi-triazoles against asexual and sexual forms of Plasmodium falciparum. Malar J 2025; 24:74. [PMID: 40038735 PMCID: PMC11881275 DOI: 10.1186/s12936-025-05297-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/15/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Despite advances in malaria chemotherapy, the disease continues to claim thousands of lives annually. Addressing this issue requires the discovery of new compounds to counteract resistance threatening the current therapeutic arsenal. In this context, bi-triazoles are substances with diverse biological activities, showing promise as lead compound to fight malaria. Triazoles are heterocyclic structures composed of five members, including three nitrogen atoms and two double bonds. Bi-triazoles, the focus of this study, are derivatives of triazoles consisting of two triazole rings (nitrogen heterocyclic) with isolated nuclei lacking a spacer and two substituents at each end. The goal of the present study was to assess the in vitro and in silico, antimalarial activity of bi-triazole compounds 14c, 14d, 13c, and 13d against asexual and sexual forms of Plasmodium falciparum. METHODS For in silico predictions, the software OSIRIS, Molinspiration, and ADMETlab were employed. To determine the 50% inhibitory concentration (IC50) on the asexual forms, the W2 clone was used, while the strain NF54 was used to assess inhibition of sexual forms. Cytotoxicity was evaluated using the HepG2 cell line, and haemolysis tests were conducted. Additionally, the selectivity index (SI) of each compound was calculated. RESULTS In silico analyses of physicochemical properties revealed that all compounds have favorable potential for drug development. Pharmacokinetics predictions also provided important, novel insights into this chemical class. Antimalarial activity tests showed that compounds 14d and 13d exhibited promising activity, with IC50 values of 3.1 and 4.4 µM, respectively. Antimalarial activity of compounds 14d and 13d may be related to the presence of methyl acetate in substituent R2 conjugated to the bi-triazole. None of the compounds demonstrated cytotoxic or haemolytic activity, with SI values above 51 for the three most active compounds, highlighting their selectivity. For the sexual forms, compounds 14c and 14d were classified as having a high potential to block malaria transmission. CONCLUSION Overall, the in vitro and in silico results showed that bi-triazole compounds may guide new biological investigation for malaria, enabling the identification and development of more active and selective antimalarial agents.
Collapse
Affiliation(s)
- Leandro do Nascimento Martinez
- Plataforma de Bioensaios de Malária E Leishmaniose (PBML), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil.
- Programa de Pós-Graduação Em Biologia Experimental (PGBIOEXP), Fundação Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil.
- Centro Universitário São Lucas -PVH/Afya, Porto Velho, RO, Brazil.
| | - Minelly Azevedo da Silva
- Instituto Federal de Educação, Ciência e Tecnologia de Rondônia - IFRO, Porto Velho, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, RO, Brazil
| | - Saara Neri Fialho
- Plataforma de Bioensaios de Malária E Leishmaniose (PBML), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
- Centro Universitário São Lucas -PVH/Afya, Porto Velho, RO, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, RO, Brazil
| | - Marcinete Latorre Almeida
- Plataforma de Bioensaios de Malária E Leishmaniose (PBML), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, RO, Brazil
| | - Amália Dos Santos Ferreira
- Plataforma de Bioensaios de Malária E Leishmaniose (PBML), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
| | - Aurileya de Jesus Gouveia
- Plataforma de Bioensaios de Malária E Leishmaniose (PBML), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
| | - Welington da Silva Paula do Nascimento
- Plataforma de Bioensaios de Malária E Leishmaniose (PBML), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, RO, Brazil
| | | | | | - Jansen Fernandes de Medeiros
- Programa de Pós-Graduação Em Biologia Experimental (PGBIOEXP), Fundação Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
- Plataforma de Infecção de Vetores da Malária (PIVEM/ Laboratório de Entomologia, Fundação Oswaldo Cruz, FIOCRUZ, UnidadeRondônia, Porto Velho, RO, Brazil
- Instituto Nacional de Epidemiologia da Amazônia Ocidental - EpiAmO, Porto Velho, RO, Brazil
| | - Natalie Ferreira Araújo
- LaSOPB - Laboratório de Síntese Orgânica e Prospecção Biológica, InstitutodeQuímica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Quelli Larissa Oliveira de Santana
- LaSOPB - Laboratório de Síntese Orgânica e Prospecção Biológica, InstitutodeQuímica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Carlos Roland Kaiser
- LaSOPB - Laboratório de Síntese Orgânica e Prospecção Biológica, InstitutodeQuímica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Sabrina Baptista Ferreira
- LaSOPB - Laboratório de Síntese Orgânica e Prospecção Biológica, InstitutodeQuímica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Maisa da Silva Araujo
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, RO, Brazil
- Plataforma de Infecção de Vetores da Malária (PIVEM/ Laboratório de Entomologia, Fundação Oswaldo Cruz, FIOCRUZ, UnidadeRondônia, Porto Velho, RO, Brazil
- Instituto Nacional de Epidemiologia da Amazônia Ocidental - EpiAmO, Porto Velho, RO, Brazil
| | - Carolina Bioni Garcia Teles
- Plataforma de Bioensaios de Malária E Leishmaniose (PBML), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
- Programa de Pós-Graduação Em Biologia Experimental (PGBIOEXP), Fundação Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
- Centro Universitário São Lucas -PVH/Afya, Porto Velho, RO, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, RO, Brazil
- Instituto Nacional de Epidemiologia da Amazônia Ocidental - EpiAmO, Porto Velho, RO, Brazil
| |
Collapse
|
10
|
Stasiak P, Sopel J, Lipowicz JM, Rawłuszko-Wieczorek AA, Korbecki J, Januchowski R. The Role of Elacridar, a P-gp Inhibitor, in the Re-Sensitization of PAC-Resistant Ovarian Cancer Cell Lines to Cytotoxic Drugs in 2D and 3D Cell Culture Models. Int J Mol Sci 2025; 26:1124. [PMID: 39940891 PMCID: PMC11817197 DOI: 10.3390/ijms26031124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Chemotherapy resistance is a significant barrier to effective cancer treatment. A key mechanism of resistance at the single-cell level is the overexpression of drug transporters in the ABC family, particularly P-glycoprotein (P-gp), which leads to multidrug resistance (MDR). Inhibitors of these transporters can help re-sensitize cancer cells to chemotherapeutics. This study evaluated elacridar (GG918 and GF120918), a potent third-generation P-gp inhibitor, for its ability to reverse MDR in paclitaxel (PAC)-resistant ovarian cancer cell lines. Sensitive and PAC-resistant cells were cultured in two-dimensional (2D) and three-dimensional (3D) models. MDR1 gene expression was analyzed using Q-PCR, and P-gp protein expression was examined via Western blot and immunofluorescence. Drug sensitivity was evaluated with MTT assays, and P-gp activity was analyzed by flow cytometry and fluorescence microscopy. Elacridar effectively inhibited P-gp activity and increased sensitivity to PAC and doxorubicin (DOX) in 2D cultures but not cisplatin (CIS). In 3D spheroids, P-gp activity inhibition was observed via Calcein-AM staining. However, no re-sensitization to PAC occurred and limited improvement was observed for DOX. These findings suggest that elacridar effectively inhibits P-gp in both 2D and 3D conditions. However, its ability to overcome drug resistance in 3D models is limited, highlighting the complexity of tissue-specific resistance mechanisms.
Collapse
Affiliation(s)
- Piotr Stasiak
- Institute of Biological Sciences, University of Zielona Góra, 65-417 Zielona Góra, Poland
- The Doctoral School of Exact and Technical Sciences, University of Zielona Góra, 65-417 Zielona Góra, Poland
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| | - Justyna Sopel
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| | - Julia Maria Lipowicz
- Department of Histology and Embryology, Doctoral School, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | | | - Jan Korbecki
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| | - Radosław Januchowski
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| |
Collapse
|
11
|
Zhang X, Guo L, Luo Y, Xu X, Han Y, Chen H, Sun H, Xue Y, Ji G. Neurotoxicity and Mechanism in Zebrafish Embryo Induced by Tetrabromobisphenol A bis (2-Hydroxyethyl) Ether (TBBPA-DHEE) Exposure. TOXICS 2025; 13:76. [PMID: 39997892 PMCID: PMC11860782 DOI: 10.3390/toxics13020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025]
Abstract
Tetrabromobisphenol A bis (2-hydroxyethyl) ether (TBBPA-DHEE), a derivative of TBBPA, has been frequently detected in the environment. In this study, the median lethal concentration (LC50) of TBBPA-DHEE at 96 h post-fertilization (hpf) was 1.573 mg/L. Based on the reported environmental concentrations, we investigated the effects of TBBPA-DHEE on the nervous system of zebrafish embryos following exposure to varying concentrations (0, 20, 100, and 500 μg/L) for 4 to 144 hpf. Our results indicated that exposure to 100 μg/L at 144 hpf led to behavioral abnormalities in zebrafish. Furthermore, exposure to TBBPA-DHEE inhibited the development of the central nervous system and motor neurons in zebrafish. Real-time polymerase chain reaction (PCR) analysis revealed that exposure to TBBPA-DHEE significantly downregulated the expression levels of neurodevelopmental genes (shha, syn2a, elavl3, gfap, and gap43). Additionally, TBBPA-DHEE increased oxidative stress in zebrafish. Transcriptomic analysis demonstrated that exposure to TBBPA-DHEE affected the signaling pathways involved in neurodevelopment. Overall, this study demonstrated that TBBPA-DHEE may disrupt the early development of the nervous system, leading to abnormal motor behavior in zebrafish larvae, and provided novel insights into the potential mechanisms of TBBPA-DHEE neurotoxicity.
Collapse
Affiliation(s)
- Xinyu Zhang
- School of Environmental Science and Engineering, Changzhou University, Changzhou 213164, China; (X.Z.); (Y.H.); (H.C.); (H.S.)
| | - Liguo Guo
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China; (L.G.); (Y.L.)
| | - Yiwen Luo
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China; (L.G.); (Y.L.)
| | - Xia Xu
- College of Urban Construction, Changzhou University, Changzhou 213164, China;
| | - Ying Han
- School of Environmental Science and Engineering, Changzhou University, Changzhou 213164, China; (X.Z.); (Y.H.); (H.C.); (H.S.)
| | - Hui Chen
- School of Environmental Science and Engineering, Changzhou University, Changzhou 213164, China; (X.Z.); (Y.H.); (H.C.); (H.S.)
| | - Haohao Sun
- School of Environmental Science and Engineering, Changzhou University, Changzhou 213164, China; (X.Z.); (Y.H.); (H.C.); (H.S.)
| | - Yingang Xue
- School of Environmental Science and Engineering, Changzhou University, Changzhou 213164, China; (X.Z.); (Y.H.); (H.C.); (H.S.)
| | - Guixiang Ji
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China; (L.G.); (Y.L.)
| |
Collapse
|
12
|
Toja-Camba FJ, Vidal-Millares M, Durán-Maseda MJ, Hermelo-Vidal G, Carracedo Á, Maroñas O, Ramudo-Cela L, Zarra-Ferro I, Fernández-Ferreiro A, Mondelo-García C. Influence of ABCB1 polymorphisms on aripiprazole and dehydroaripiprazole plasma concentrations. Sci Rep 2025; 15:1521. [PMID: 39789135 PMCID: PMC11717999 DOI: 10.1038/s41598-024-84192-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Aripiprazole (ARI) is an atypical antipsychotic which is a substrate of P-glycoprotein (P-gp), a transmembrane glycoprotein that plays a crucial role in eliminating potentially harmful compounds from the organism. ARI once-monthly (AOM) is a long-acting injectable form which improves treatment compliance. Genetic polymorphisms in ABCB1 may lead to changes in P-gp function, leading to individual differences in drug disposition. The present study aims to determine how the different variants of the three most prevalent SNPs of the ABCB1 gene affect plasma concentrations of ARI, of its active metabolite dehydroaripiprazole (DHA) and ARI/DHA ratio in patients under AOM treatment. The metabolizing state of the two main aripiprazole metabolizing enzymes (CYP2D6 and CYP3A4) were considered to specifically study the effect of P-gp on plasma concentrations of the parent compound and its active metabolite. The study found a clear relationship between the genotypes found for the different ABCB1 SNPs and the ARI/DHA ratio. Specifically, patients with GG genotype in G2677T have almost twice the ratio compared to TT genotype. Similarly, this increase is also found in C3435T with 1.4-fold and in C1236T with 1.6-fold for the same genotypes. Regarding haplotypes, significant differences were obtained between CC-GG-CC and TT-TT-TT patients, with an 87.9% higher ratio in patients with the CC-GG-CC haplotype. There was a clear trend towards lower ARI concentrations and higher DHA concentrations when the presence of mutated T alleles increases. The ABCB1 gene could be a good partner along with CYP2D6 and CYP3A4 genotyping in conjunction with monitoring ARI plasma concentrations.
Collapse
Affiliation(s)
- Francisco José Toja-Camba
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706, Santiago de Compostela, Spain
- FarmaCHUSLab Group, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
- Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - María Vidal-Millares
- Psychiatry Department, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - María José Durán-Maseda
- Psychiatry Department, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Gonzalo Hermelo-Vidal
- FarmaCHUSLab Group, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Ángel Carracedo
- Genetics Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Galician Foundation of Genomic Medicine, Foundation of Health Research Institute of Santiago de Compostela (FIDIS), SERGAS, Santiago de Compostela, Spain
- Centre for Biomedical Network Research On Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain
| | - Olalla Maroñas
- Galician Foundation of Genomic Medicine, Foundation of Health Research Institute of Santiago de Compostela (FIDIS), SERGAS, Santiago de Compostela, Spain
- Centre for Biomedical Network Research On Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain
- Pharmacogenomics and Drug Discovery (GenDeM), Foundation of Health Research Institute of Santiago de Compostela (FIDIS), Galicia, Santiago de Compostela, Spain
| | - Luis Ramudo-Cela
- Pharmacy Department, University Clinical Hospital A Coruña (CHUAC), A Coruña, Spain
- Hospital Pharmacy Research Group, Health Research Institute of A Coruña (INIBIC), A Coruña, Spain
| | - Irene Zarra-Ferro
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706, Santiago de Compostela, Spain
- FarmaCHUSLab Group, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Anxo Fernández-Ferreiro
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706, Santiago de Compostela, Spain.
- FarmaCHUSLab Group, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain.
| | - Cristina Mondelo-García
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706, Santiago de Compostela, Spain.
- FarmaCHUSLab Group, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain.
| |
Collapse
|
13
|
Rahman I, Liang B, Sajid A, Ambudkar SV, Huang H. Photodynamic priming modulates cellular ATP levels to overcome P-glycoprotein-mediated drug efflux in chemoresistant triple-negative breast cancer. Photochem Photobiol 2025; 101:188-205. [PMID: 38824410 PMCID: PMC11737009 DOI: 10.1111/php.13970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 06/03/2024]
Abstract
P-glycoprotein (P-gp, ABCB1) is a well-researched ATP-binding cassette (ABC) drug efflux transporter linked to the development of cancer multidrug resistance (MDR). Despite extensive studies, approved therapies to safely inhibit P-gp in clinical settings are lacking, necessitating innovative strategies beyond conventional inhibitors or antibodies to reverse MDR. Photodynamic therapy is a globally approved cancer treatment that uses targeted, harmless red light to activate non-toxic photosensitizers, confining its cytotoxic photochemical effects to disease sites while sparing healthy tissues. This study demonstrates that photodynamic priming (PDP), a sub-cytotoxic photodynamic therapy process, can inhibit P-gp function by modulating cellular respiration and ATP levels in light accessible regions. Using chemoresistant (VBL-MDA-MB-231) and chemosensitive (MDA-MB-231) triple-negative breast cancer cell lines, we showed that PDP decreases mitochondrial membrane potential by 54.4% ± 30.4 and reduces mitochondrial ATP production rates by 94.9% ± 3.46. Flow cytometry studies showed PDP can effectively improve the retention of P-gp substrates (calcein) by up to 228.4% ± 156.3 in chemoresistant VBL-MDA-MB-231 cells, but not in chemosensitive MDA-MB-231 cells. Further analysis revealed that PDP did not alter the cell surface expression level of P-gp in VBL-MDA-MB-231 cells. These findings indicate that PDP can reduce cellular ATP below the levels that is required for the function of P-gp and improve intracellular substrate retention. We propose that PDP in combination with chemotherapy drugs, might improve the efficacy of chemotherapy and overcome cancer MDR.
Collapse
Affiliation(s)
- Idrisa Rahman
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMarylandUSA
- Laboratory of Cell Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Barry Liang
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMarylandUSA
- Laboratory of Cell Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Andaleeb Sajid
- Laboratory of Cell Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Suresh V. Ambudkar
- Laboratory of Cell Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Huang‐Chiao Huang
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMarylandUSA
| |
Collapse
|
14
|
Yong-Gonzalez V, Radu C, Calder PA, Shum D, Gin DY, Frattini MG, Djaballah H. Antiproliferative Activity of Cephalotaxus Esters: Overcoming Chemoresistance. Comb Chem High Throughput Screen 2025; 28:1264-1275. [PMID: 39444179 DOI: 10.2174/0113862073322175240823104921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/05/2024] [Accepted: 07/18/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Omacetaxine, a semisynthetic form of Homoharringtonine (HHT), was approved for the treatment of Chronic Myeloid Leukemia (CML). Previously, we have published the synthesis of this natural alkaloid and three of its derivatives: Deoxyharringtonine (DHT), Deoxyhomoharringtonine (DHHT), and Bis(demethyl)-deoxyharringtonine (BDHT), and reported its refractory activity against the HL-60/RV+ cells over-expressing P-glycoprotein 1 (MDR1). METHODS In this study, we have explored the extent of this resistance by first expanding the panel of established cell lines and using a panel of 21 leukemia patient-derived primary cells. RESULTS Herein, we have reported consistent resistance to HTT of K562-derived cells and to mitoxantrone of MES-SA/MX2-derived cells; all of them have been found to overexpress MDR1, while we have found U87MG-ABCG2 and H69AR cells to be very sensitive to HTT. In contrast, DHT, DHHT, and BDHT seemingly overcame this resistance due to the changes made to the acyl chain of HTT, rendering the derivatives less susceptible to efflux. Surprisingly, the leukemia primary cells were very sensitive to HHT and its derivatives with low nanomolar potencies, followed by a new class of CDC7 kinase inhibitors, the anthracycline class of topoisomerase inhibitors, the DNA intercalator actinomycin-D, and the vinca alkaloid class of microtubule inhibitors. The mechanism of cell death induced by HTT and DHHT was found to be mediated via caspase 3 cleavage, leading to apoptosis. CONCLUSION Taken together, our results confirm that HHT is a substrate for MDR1. It opens the door to a new opportunity to clinically evaluate HHT and its derivatives for the treatment of AML and other cancers.
Collapse
Affiliation(s)
- Vladimir Yong-Gonzalez
- HTS Core Facility - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Department of Analytical Chemistry, C16 Biosciences, New York, NY 10019, USA
| | - Constantin Radu
- HTS Core Facility - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Automation and Workflow Solutions, Molecular Devices, San Jose, CA 95134, USA
| | - Paul A Calder
- HTS Core Facility - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Shum
- HTS Core Facility - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Screening Discovery Platform, Institut Pasteur Korea, Seongnamsi, Gyeonggido, 13488, Republic of Korea
| | - David Y Gin
- Molecular Pharmacology Program - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Deceased
| | - Mark G Frattini
- Department of Medicine - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Office of the CMO, Cellectis Inc, New York, NY 10016, USA
| | - Hakim Djaballah
- HTS Core Facility - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Molecular Pharmacology Program - Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Office of the CEO, Keren Therapeutics, New York, NY 10583, USA
| |
Collapse
|
15
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2025; 45:311-343. [PMID: 39180410 DOI: 10.1002/med.22077] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
16
|
Lo Buglio G, Lo Cicero A, Campora S, Ghersi G. The Multifaced Role of Collagen in Cancer Development and Progression. Int J Mol Sci 2024; 25:13523. [PMID: 39769286 PMCID: PMC11678882 DOI: 10.3390/ijms252413523] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/05/2025] Open
Abstract
Collagen is a crucial protein in the extracellular matrix (ECM) essential for preserving tissue architecture and supporting crucial cellular functions like proliferation and differentiation. There are twenty-eight identified types of collagen, which are further divided into different subgroups. This protein plays a critical role in regulating tissue homeostasis. However, in solid tumors, the balance can be disrupted, due to an abundance of collagen in the tumor microenvironment, which significantly affects tumor growth, cell invasion, and metastasis. It is important to investigate the specific types of collagens in cancer ECM and their distinct roles in tumor progression to comprehend their unique contribution to tumor behavior. The diverse pathophysiological functions of different collagen types in cancers illustrate collagen's dual roles, offering potential therapeutic options and serving as prognostic markers.
Collapse
Affiliation(s)
- Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Abiel srl, 90128 Palermo, Italy
| |
Collapse
|
17
|
Elbahnsi A, Dudas B, Cisternino S, Declèves X, Miteva MA. Mechanistic insights into P-glycoprotein ligand transport and inhibition revealed by enhanced molecular dynamics simulations. Comput Struct Biotechnol J 2024; 23:2548-2564. [PMID: 38989058 PMCID: PMC11233806 DOI: 10.1016/j.csbj.2024.06.010] [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: 05/02/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
P-glycoprotein (P-gp) plays a crucial role in cellular detoxification and drug efflux processes, transitioning between inward-facing (IF) open, occluded, and outward-facing (OF) states to facilitate substrate transport. Its role is critical in cancer therapy, where P-gp contributes to the multidrug resistance phenotype. In our study, classical and enhanced molecular dynamics (MD) simulations were conducted to dissect the structural and functional features of the P-gp conformational states. Our advanced MD simulations, including kinetically excited targeted MD (ketMD) and adiabatic biasing MD (ABMD), provided deeper insights into state transition and translocation mechanisms. Our findings suggest that the unkinking of TM4 and TM10 helices is a prerequisite for correctly achieving the outward conformation. Simulations of the IF-occluded conformations, characterized by kinked TM4 and TM10 helices, consistently demonstrated altered communication between the transmembrane domains (TMDs) and nucleotide binding domain 2 (NBD2), suggesting the implication of this interface in inhibiting P-gp's efflux function. A particular emphasis was placed on the unstructured linker segment connecting the NBD1 to TMD2 and its role in the transporter's dynamics. With the linker present, we specifically noticed a potential entrance of cholesterol (CHOL) through the TM4-TM6 portal, shedding light on crucial residues involved in accommodating CHOL. We therefore suggest that this entry mechanism could be employed for some P-gp substrates or inhibitors. Our results provide critical data for understanding P-gp functioning and developing new P-gp inhibitors for establishing more effective strategies against multidrug resistance.
Collapse
Affiliation(s)
- Ahmad Elbahnsi
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, Paris, France
| | - Balint Dudas
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, Paris, France
| | - Salvatore Cisternino
- Université Paris Cité, Inserm UMRS 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Xavier Declèves
- Université Paris Cité, Inserm UMRS 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Maria A. Miteva
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, Paris, France
| |
Collapse
|
18
|
Dey Bhowmik A, Shaw P, Gopinatha Pillai MS, Rao G, Dwivedi SKD. Evolving landscape of detection and targeting miRNA/epigenetics for therapeutic strategies in ovarian cancer. Cancer Lett 2024; 611:217357. [PMID: 39615646 PMCID: PMC12119979 DOI: 10.1016/j.canlet.2024.217357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
Ovarian cancer (OC) accounts for the highest mortality rates among all gynecologic malignancies. The high mortality of OC is often associated with delayed detection, prolonged latency, enhanced metastatic potential, acquired drug resistance, and frequent recurrence. This review comprehensively explores key aspects of OC, including cancer diagnosis, mechanisms of disease resistance, and the pivotal role of epigenetic regulation, particularly by microRNAs (miRs) in cancer progression. We highlight the intricate regulatory mechanisms governing miR expression within the context of OC and the current status of epigenetic advancement in the therapeutic development and clinical trial progression. Through network analysis we elucidate the regulatory interactions between dysregulated miRs in OC and their targets which are involved in different signaling pathways. By exploring these interconnected facets and critical analysis, we endeavor to provide a nuanced understanding of the molecular dynamics underlying OC, its detection and shedding light on potential avenues for miRs and epigenetics-based therapeutic intervention and management strategies.
Collapse
Affiliation(s)
- Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
19
|
Sinanian MM, Rahman A, Elshazly AM, Neely V, Nagarajan B, Kellogg GE, Risinger AL, Gewirtz DA. A BPTF Inhibitor That Interferes with the Multidrug Resistance Pump to Sensitize Murine Triple-Negative Breast Cancer Cells to Chemotherapy. Int J Mol Sci 2024; 25:11346. [PMID: 39518898 PMCID: PMC11545213 DOI: 10.3390/ijms252111346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/31/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with a generally poor prognosis due to its highly aggressive and metastatic nature, lack of targetable receptors, as well as the frequent development of resistance to chemotherapy. We previously reported that AU1, a small molecule developed as an inhibitor of BPTF (bromodomain PHD finger-containing transcription factor), was capable of sensitizing preclinical models of TNBC to chemotherapy in part via the promotion of autophagy. In studies reported here, we identify an additional property of this compound, specifically that sensitization is associated with the inhibition of the P-glycoprotein (P-gp) efflux pump. In silico molecular docking studies indicate that AU1 binds to active regions of the efflux pump in a manner consistent with the inhibition of the pump function. This work identifies a novel chemical structure that can influence multidrug efflux, an established mechanism of drug resistance in TNBC, that has not yet been successfully addressed by clinical efforts.
Collapse
Affiliation(s)
- Melanie M. Sinanian
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
| | - Afshan Rahman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
| | - Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Victoria Neely
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Balaji Nagarajan
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA; (B.N.); (G.E.K.)
| | - Glen E. Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA; (B.N.); (G.E.K.)
| | - April L. Risinger
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229, USA;
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
| |
Collapse
|
20
|
Yao Y, Fan D. Advances in MUC1 resistance to chemotherapy in pancreatic cancer. J Chemother 2024; 36:449-456. [PMID: 38006297 DOI: 10.1080/1120009x.2023.2282839] [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: 07/26/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
The incidence of pancreatic cancer (PC), a highly fatal malignancy, is increasing every year. Chemotherapy is an important treatment for it in addition to surgery, yet most patients become resistant to chemotherapeutic agents within a few weeks of treatment initiation. MUC1 is a highly glycosylated transmembrane protein, and studies have shown that aberrantly glycosylated overexpression of MUC1 is involved in regulating the biology of chemoresistance in cancer cells. This article summarizes the mechanism of MUC1 in PC chemoresistance and reviews MUC1-based targeted therapies.
Collapse
Affiliation(s)
- Youhao Yao
- The Fifth Clinical Medical College of Shanxi Medical University, Shanxi, PR China
- Surgery Department, Shanxi Provincial People's Hospital, Taiyuan, PR China
| | - Daguang Fan
- Surgery Department, Shanxi Provincial People's Hospital, Taiyuan, PR China
| |
Collapse
|
21
|
Roucairol M, Georgiou A, Cazenave T, Prischi F, Pardo OE. DrugSynthMC: An Atom-Based Generation of Drug-like Molecules with Monte Carlo Search. J Chem Inf Model 2024; 64:7097-7107. [PMID: 39249497 PMCID: PMC11423341 DOI: 10.1021/acs.jcim.4c01451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
A growing number of deep learning (DL) methodologies have recently been developed to design novel compounds and expand the chemical space within virtual libraries. Most of these neural network approaches design molecules to specifically bind a target based on its structural information and/or knowledge of previously identified binders. Fewer attempts have been made to develop approaches for de novo design of virtual libraries, as synthesizability of generated molecules remains a challenge. In this work, we developed a new Monte Carlo Search (MCS) algorithm, DrugSynthMC (Drug Synthesis using Monte Carlo), in conjunction with DL and statistical-based priors to generate thousands of interpretable chemical structures and novel drug-like molecules per second. DrugSynthMC produces drug-like compounds using an atom-based search model that builds molecules as SMILES, character by character. Designed molecules follow Lipinski's "rule of 5″, show a high proportion of highly water-soluble nontoxic predicted-to-be synthesizable compounds, and efficiently expand the chemical space within the libraries, without reliance on training data sets, synthesizability metrics, or enforcing during SMILES generation. Our approach can function with or without an underlying neural network and is thus easily explainable and versatile. This ease in drug-like molecule generation allows for future integration of score functions aimed at different target- or job-oriented goals. Thus, DrugSynthMC is expected to enable the functional assessment of large compound libraries covering an extensive novel chemical space, overcoming the limitations of existing drug collections. The software is available at https://github.com/RoucairolMilo/DrugSynthMC.
Collapse
Affiliation(s)
- Milo Roucairol
- LAMSADE, Université Paris-Dauphine, Pl. du Maréchal de Lattre de Tassigny, 75016 Paris, France
| | - Alexios Georgiou
- LAMSADE, Université Paris-Dauphine, Pl. du Maréchal de Lattre de Tassigny, 75016 Paris, France
| | - Tristan Cazenave
- LAMSADE, Université Paris-Dauphine, Pl. du Maréchal de Lattre de Tassigny, 75016 Paris, France
| | - Filippo Prischi
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London SE1 1UL, United Kingdom
| | - Olivier E Pardo
- Division of Cancer, Department of Surgery and Cancer, Imperial College, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
22
|
McCormick LA, McCormick JW, Park C, Follit CA, Wise JG, Vogel PD. Computationally accelerated identification of P-glycoprotein inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583428. [PMID: 39345515 PMCID: PMC11430104 DOI: 10.1101/2024.03.05.583428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Overexpression of the polyspecific efflux transporter, P-glycoprotein (P-gp, MDR1, ABCB1 ), is a major mechanism by which cancer cells acquire multidrug resistance (MDR), the resistance to diverse chemotherapeutic drugs. Inhibiting drug transport by P-gp can resensitize cancer cells to chemotherapy, but there are no P-gp inhibitors available to patients. Clinically unsuccessful P-gp inhibitors tend to bind at the pump's transmembrane drug binding domains and are often P-gp transport substrates, resulting in lowered intracellular concentration of the drug and altered pharmacokinetics. In prior work, we used computationally accelerated drug discovery to identify novel P-gp inhibitors that target the pump's cytoplasmic nucleotide binding domains. Our first-draft study provided conclusive evidence that the nucleotide binding domains of P-gp are viable targets for drug discovery. Here we develop an enhanced, computationally accelerated drug discovery pipeline that expands upon our prior work by iteratively screening compounds against multiple conformations of P-gp with molecular docking. Targeted molecular dynamics simulations with our homology model of human P-gp were used to generate docking receptors in conformations mimicking a putative drug transport cycle. We offset the increased computational complexity using custom Tanimoto chemical datasets, which maximize the chemical diversity of ligands screened by docking. Using our expanded, virtual-assisted pipeline, we identified nine novel P-gp inhibitors that reverse MDR in two types of P-gp overexpressing human cancer cell lines, reflecting a 13.4% hit rate. Of these inhibitors, all were non-toxic to non-cancerous human cells, and six were not likely to be transport substrates of P-gp. Our novel P-gp inhibitors are chemically diverse and are good candidates for lead optimization. Our results demonstrate that the nucleotide binding domains of P-gp are an underappreciated target in the effort to reverse P-gp-mediated multidrug resistance in cancer.
Collapse
|
23
|
Ouellette V, Bouzriba C, Chavez Alvarez AC, Bruxelles Q, Hamel-Côté G, Fortin S. Pyridinyl 4-(2-oxoalkylimidazolidin-1-yl)benzenesulfonates and their hydrochloride salts as novel water soluble antimitotic prodrugs bioactivated by cytochrome P450 1A1 in breast cancer cells. RSC Med Chem 2024:d4md00476k. [PMID: 39281801 PMCID: PMC11393734 DOI: 10.1039/d4md00476k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024] Open
Abstract
We developed first-in-class antimitotic prodrugs phenyl 4-(2-oxo-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) bioactivated by cytochrome P450 (CYP) 1A1 that are highly selective toward several breast cancer cells. However, they show sparingly water solubility. Therefore, we replaced their phenyl ring B with a substituted pyridinyl group preparing novel pyridinyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PYRAIB-SOs) and their hydrochloride salts. Our results evidence that PYRAIB-SO hydrochloride salts show higher water solubility compared to their neutral and PAIB-SO counterparts by up to 625-fold. PYRAIB-SOs with a nitrogen atom at position 3 of the pyridinyl ring exhibited strong antiproliferative activity (IC50: 0.03-3.3 μM) and high selectivity (8->1250) toward sensitive CYP1A1-positive breast cancer cells and cells stably transfected with CYP1A1. They induce cell cycle arrest in the G2/M phase and disrupt microtubule dynamic assembly. Enzymatic assays confirmed that CYP1A1 metabolizes PYRAIB-SOs into their active form with in vitro hepatic half-lives (55-120 min) in rodent and human liver microsomes. Overall, this will allow to increase drug concentration for in vivo studies.
Collapse
Affiliation(s)
- Vincent Ouellette
- Hôpital Saint-François d'Assise, Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie 10 Rue de l'Espinay Québec QC G1L 3L5 Canada (418) 525 4444 ext. 52364
- Faculté de pharmacie, Université Laval Québec QC G1V 0A6 Canada
| | - Chahrazed Bouzriba
- Hôpital Saint-François d'Assise, Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie 10 Rue de l'Espinay Québec QC G1L 3L5 Canada (418) 525 4444 ext. 52364
- Faculté de pharmacie, Université Laval Québec QC G1V 0A6 Canada
| | - Atziri Corin Chavez Alvarez
- Hôpital Saint-François d'Assise, Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie 10 Rue de l'Espinay Québec QC G1L 3L5 Canada (418) 525 4444 ext. 52364
- Faculté de pharmacie, Université Laval Québec QC G1V 0A6 Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval 2725 Ch Ste-Foy Québec QC G1V 4G5 Canada
| | - Quentin Bruxelles
- Hôpital Saint-François d'Assise, Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie 10 Rue de l'Espinay Québec QC G1L 3L5 Canada (418) 525 4444 ext. 52364
- Faculté de pharmacie, Université Laval Québec QC G1V 0A6 Canada
| | - Geneviève Hamel-Côté
- Hôpital Saint-François d'Assise, Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie 10 Rue de l'Espinay Québec QC G1L 3L5 Canada (418) 525 4444 ext. 52364
| | - Sébastien Fortin
- Hôpital Saint-François d'Assise, Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie 10 Rue de l'Espinay Québec QC G1L 3L5 Canada (418) 525 4444 ext. 52364
- Faculté de pharmacie, Université Laval Québec QC G1V 0A6 Canada
| |
Collapse
|
24
|
Ebrahimi S, Khaleghi Ghadiri M, Stummer W, Gorji A. Enhancing 5-ALA-PDT efficacy against resistant tumor cells: Strategies and advances. Life Sci 2024; 351:122808. [PMID: 38852796 DOI: 10.1016/j.lfs.2024.122808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
As a precursor of protoporphyrin IX (PpIX), an endogenous pro-apoptotic and fluorescent molecule, 5-Aminolevulinic acid (5-ALA) has gained substantial attention for its potential in fluorescence-guided surgery as well as photodynamic therapy (PDT). Moreover, 5-ALA-PDT has been suggested as a promising chemo-radio sensitization therapy for various cancers. However, insufficient 5-ALA-induced PpIX fluorescence and the induction of multiple resistance mechanisms may hinder the 5-ALA-PDT clinical outcome. Reduced efficacy and resistance to 5-ALA-PDT can result from genomic alterations, tumor heterogeneity, hypoxia, activation of pathways related to cell surveillance, production of nitric oxide, and most importantly, deregulated 5-ALA transporter proteins and heme biosynthesis enzymes. Understanding the resistance regulatory mechanisms of 5-ALA-PDT may allow the development of effective personalized cancer therapy. Here, we described the mechanisms underlying resistance to 5-ALA-PTD across various tumor types and explored potential strategies to overcome this resistance. Furthermore, we discussed future approaches that may enhance the efficacy of treatments using 5-ALA-PDT.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Epilepsy Research Center, Münster University, 48149 Münster, Germany; Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran
| | | | - Walter Stummer
- Department of Neurosurgery, Münster University, 48149 Münster, Germany
| | - Ali Gorji
- Epilepsy Research Center, Münster University, 48149 Münster, Germany; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; Neuroscience Research Center, Mashhad University of Medical Sciences, 9177948564 Mashhad, Iran.
| |
Collapse
|
25
|
Song W, Yang H, Wang Y, Chen S, Zhong W, Wang Q, Ding W, Xu G, Meng C, Liang Y, Chen Z, Cao S, Wei L, Li F. Glutathione-Sensitive Photosensitizer-Drug Conjugates Target the Mitochondria to Overcome Multi-Drug Resistance in Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307765. [PMID: 38898730 PMCID: PMC11321625 DOI: 10.1002/advs.202307765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 05/11/2024] [Indexed: 06/21/2024]
Abstract
Multi-drug resistance (MDR) is a major cause of cancer therapy failure. Photodynamic therapy (PDT) is a promising modality that can circumvent MDR and synergize with chemotherapies, based on the generation of reactive oxygen species (ROS) by photosensitizers. However, overproduction of glutathione (GSH) by cancer cells scavenges ROS and restricts the efficacy of PDT. Additionally, side effects on normal tissues are unavoidable after PDT treatment. Here, to develop organic systems that deliver effective anticancer PDT and chemotherapy simultaneously with very little side effects, three GSH-sensitive photosensitizer-drug conjugates (CyR-SS-L) are designed and synthesized. CyR-SS-L localized in the mitochondria then is cleaved into CyR-SG and SG-L parts by reacting with and consuming high levels of intracellular GSH. Notably, CyR-SG generates high levels of ROS in tumor cells instead of normal cells and be exploited for PDT and the SG-L part is used for chemotherapy. CyR-SS-L inhibits better MDR cancer tumor inhibitory activity than indocyanine green, a photosensitizer (PS) used for PDT in clinical applications. The results appear to be the first to show that CyR-SS-L may be used as an alternative PDT agent to be more effective against MDR cancers without obvious damaging normal cells by the combination of PDT, GSH depletion, and chemotherapy.
Collapse
Affiliation(s)
- Weiguo Song
- Department of Medicinal ChemistrySchool of PharmacyShandong UniversityJinan250012China
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Hekai Yang
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Ying Wang
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Shuzhen Chen
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Wenda Zhong
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Qian Wang
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Wenshuo Ding
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Guangzhao Xu
- Weifang Synovtech New Material Technology CO., LTD.Weifang262700China
- Harway Pharma Co., Ltd.Dongying254753China
| | - Chen Meng
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Ying Liang
- Department of General PracticeThe First Affiliated Hospital of Shandong First Medical UniversityJinan250013China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Shuhua Cao
- College of ChemistryChemical and Environmental EngineeringWeifang UniversityWeifang261061China
| | - Liuya Wei
- School of PharmacyWeifang Medical UniversityWeifang261053China
| | - Fahui Li
- School of PharmacyWeifang Medical UniversityWeifang261053China
| |
Collapse
|
26
|
Chembukavu SN, Lindsay AJ. Therapy-induced senescence in breast cancer: an overview. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:902-920. [PMID: 39280248 PMCID: PMC11390292 DOI: 10.37349/etat.2024.00254] [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: 04/04/2024] [Accepted: 06/06/2024] [Indexed: 09/18/2024] Open
Abstract
Outcomes for women with breast cancer have improved dramatically in recent decades. However, many patients present with intrinsic drug resistance and others are initially sensitive to anti-cancer drugs but acquire resistance during the course of their treatment, leading to recurrence and/or metastasis. Drug therapy-induced senescence (TIS) is a form of drug resistance characterised by the induction of cell cycle arrest and the emergence of a senescence-associated secretory phenotype (SASP) that can develop in response to chemo- and targeted- therapies. A wide range of anticancer interventions can lead to cell cycle arrest and SASP induction, by inducing genotoxic stress, hyperactivation of signalling pathways or oxidative stress. TIS can be anti-tumorigenic in the short-term, but pro-tumorigenic in the long-term by creating a pro-inflammatory and immunosuppressive microenvironment. Moreover, the SASP can promote angiogenesis and epithelial-mesenchymal transition in neighbouring cells. In this review, we will describe the characteristics of TIS in breast cancer and detail the changes in phenotype that accompany its induction. We also discuss strategies for targeting senescent cancer cells in order to prevent or delay tumour recurrence.
Collapse
Affiliation(s)
- Suraj Narayanan Chembukavu
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| | - Andrew J Lindsay
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| |
Collapse
|
27
|
Tufail M, Hu JJ, Liang J, He CY, Wan WD, Huang YQ, Jiang CH, Wu H, Li N. Hallmarks of cancer resistance. iScience 2024; 27:109979. [PMID: 38832007 PMCID: PMC11145355 DOI: 10.1016/j.isci.2024.109979] [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: 06/05/2024] Open
Abstract
This review explores the hallmarks of cancer resistance, including drug efflux mediated by ATP-binding cassette (ABC) transporters, metabolic reprogramming characterized by the Warburg effect, and the dynamic interplay between cancer cells and mitochondria. The role of cancer stem cells (CSCs) in treatment resistance and the regulatory influence of non-coding RNAs, such as long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), are studied. The chapter emphasizes future directions, encompassing advancements in immunotherapy, strategies to counter adaptive resistance, integration of artificial intelligence for predictive modeling, and the identification of biomarkers for personalized treatment. The comprehensive exploration of these hallmarks provides a foundation for innovative therapeutic approaches, aiming to navigate the complex landscape of cancer resistance and enhance patient outcomes.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Ju Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Liang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Cai-Yun He
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Qi Huang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Bondock S, Alabbad N, Hossan A, Abdou MM, Shati AA, Alfaifi MY, Elbehairi SEI, Mohamed NM. Design, synthesis, and anticancer evaluation of novel coumarin/thiazole congeners as potential CDK2 inhibitors with molecular dynamics. RSC Adv 2024; 14:18838-18855. [PMID: 38873551 PMCID: PMC11167341 DOI: 10.1039/d4ra02456g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
A series of novel coumarin-thiazoles was designed and synthesized as a possible CDK2 inhibitor with anticancer activity with low toxicity. The design relied on having hydrazine thiazole or its open-form thioamide to form H-bonds with the ATP binding site while coumarin maintained the crucial hydrophobic interactions for proper fitting. The biological evaluation revealed that the hydroxycoumarin-thiazole derivative 6c demonstrated the best inhibition with HepG2 and HCT116 IC50 2.6 and 3.5 μM, respectively. Similarly, its open thioamide chain congener 5c exhibited potent inhibition on MCF-7 and HepG2 with IC50 of 4.5 and 5.4 μM, respectively. Molecular docking simulations supported the assumption of inhibiting CDK2 by preserving the crucial interaction pattern with the hinge ATP site and the surrounding hydrophobic (HPO) side chains. Furthermore, molecular dynamics simulations of 5c and 6c established satisfactory stability and affinity within the CDK2 active site.
Collapse
Affiliation(s)
- Samir Bondock
- Chemistry Department, Faculty of Science, King Khalid University 9004 Abha Saudi Arabia
| | - Nada Alabbad
- Chemistry Department, Faculty of Science, King Khalid University 9004 Abha Saudi Arabia
| | - Aisha Hossan
- Chemistry Department, Faculty of Science, King Khalid University 9004 Abha Saudi Arabia
| | - Moaz M Abdou
- Egyptian Petroleum Research Institute Nasr City 11727 Cairo Egypt
| | - Ali A Shati
- Biology Department, Faculty of Science, King Khalid University 9004 Abha Saudi Arabia
| | - Mohammad Y Alfaifi
- Biology Department, Faculty of Science, King Khalid University 9004 Abha Saudi Arabia
| | - Serag E I Elbehairi
- Biology Department, Faculty of Science, King Khalid University 9004 Abha Saudi Arabia
| | - Nada M Mohamed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information, MTI Cairo Egypt
| |
Collapse
|
29
|
Gadwal A, Purohit P, Khokhar M, Vishnoi JR, Pareek P, Choudhary R, Elhence P, Banerjee M, Sharma P. GALNT14 in association with GDF-15 promotes stemness and drug resistance through β-catenin signalling pathway in breast cancer. Mol Biol Rep 2024; 51:691. [PMID: 38796671 DOI: 10.1007/s11033-024-09645-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/16/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Altered glycosylation plays a role in carcinogenesis. GALNT14 promotes cancer stem-like properties and drug resistance. GDF-15 is known to induces drug resistance and stemness markers for maintenance of breast cancer (BC) stem-like cell state. Currently there is lack of data on association of GDF-15 and GALNTs. In this study, the expression and interaction of GALNT14 and GDF-15 with stemness (OCT4 and SOX2) and drug resistance (ABCC5) markers were evaluated in BC. METHODS We investigated tumour tissue from 30 BC patients and adjacent non-tumour tissues. Expression of serum GALNT14 from BC patients and matched healthy controls was evaluated. Expression of GALNT14, GDF-15, OCT4, SOX2, ABCC5, and β-catenin in BC tissue was determined by RT-PCR. Knockdown of GALNT14 and GDF-15 in the MCF-7 cell line was done through siRNA, gene expression and protein expression of β-catenin by western blot were determined. RESULTS A significant increase in the expression of GALNT14, GDF-15, OCT4, SOX2, ABCC5, and β-catenin was observed in BC tumour tissues compared to adjacent non-tumour tissues. The serum level of GALNT14 was significantly high in BC patients (80.7 ± 65.3 pg/ml) compared to healthy controls (12.2 ± 9.12 pg/ml) (p < 0.000). To further analyse the signalling pathway involved in BC stemness and drug resistance, GALNT14 and GDF-15 were knocked down in the MCF-7 cell line, and it was observed that after knockdown, the expression level of OCT4, SOX2, ABCC5, and β-catenin was decreased, and co-knockdown with GALNT14 and GDF-15 further decreased the expression of genes. CONCLUSION It can be concluded that GALNT14, in association with GDF-15, promotes stemness and intrinsic drug resistance in BC, possibly through the β-catenin signalling pathway.
Collapse
Affiliation(s)
- Ashita Gadwal
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India.
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Jeewan Ram Vishnoi
- Department of Oncosurgery, All India Institute of Medical Sciences, Jodhpur, India
| | - Puneet Pareek
- Department of Radiation Oncology, All India Institute of Medical Sciences, Jodhpur, India
| | - Ramkaran Choudhary
- Department of General Surgery, All India Institute of Medical Sciences, Jodhpur, India
| | - Poonam Elhence
- Department of Pathology, All India Institute of Medical Sciences, Jodhpur, India
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| |
Collapse
|
30
|
Mughis H, Lye P, Imperio GE, Bloise E, Matthews SG. Hypoxia modulates P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) drug transporters in brain endothelial cells of the developing human blood-brain barrier. Heliyon 2024; 10:e30207. [PMID: 38737275 PMCID: PMC11088273 DOI: 10.1016/j.heliyon.2024.e30207] [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: 07/20/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) multidrug resistance (MDR) transporters are localized at the luminal surface of the blood-brain barrier (BBB). They confer fetal brain protection against harmful compounds that may be circulating in the peripheral blood. The fetus develops in low oxygen levels; however, some obstetric pathologies such as pre-eclampsia, placenta accreta/previa may result in even greater fetal hypoxic states. We investigated how hypoxia impacts MDR transporters in human fetal brain endothelial cells (hfBECs) derived from early and mid-stages of pregnancy. Hypoxia decreased BCRP protein and activity in hfBECs derived in early pregnancy. In contrast, in hfBECs derived in mid-pregnancy there was an increase in P-gp and BCRP activity following hypoxia. Results suggest a hypoxia-induced reduction in fetal brain protection in early pregnancy, but a potential increase in transporter-mediated protection at the BBB during mid-gestation. This would modify accumulation of various key physiological and pharmacological substrates of P-gp and BCRP in the developing fetal brain and potentially contribute to the pathogenesis of neurodevelopmental disorders commonly associated with in utero hypoxia.
Collapse
Affiliation(s)
- Hafsah Mughis
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Phetcharawan Lye
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Guinever E. Imperio
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Enrrico Bloise
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Departmento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Stephen G. Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Obstetrics & Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Svane N, Pedersen ABV, Rodenberg A, Ozgür B, Saaby L, Bundgaard C, Kristensen M, Tfelt-Hansen P, Brodin B. The putative proton-coupled organic cation antiporter is involved in uptake of triptans into human brain capillary endothelial cells. Fluids Barriers CNS 2024; 21:39. [PMID: 38711118 DOI: 10.1186/s12987-024-00544-6] [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: 03/13/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Triptans are anti-migraine drugs with a potential central site of action. However, it is not known to what extent triptans cross the blood-brain barrier (BBB). The aim of this study was therefore to determine if triptans pass the brain capillary endothelium and investigate the possible underlying mechanisms with focus on the involvement of the putative proton-coupled organic cation (H+/OC) antiporter. Additionally, we evaluated whether triptans interacted with the efflux transporter, P-glycoprotein (P-gp). METHODS We investigated the cellular uptake characteristics of the prototypical H+/OC antiporter substrates, pyrilamine and oxycodone, and seven different triptans in the human brain microvascular endothelial cell line, hCMEC/D3. Triptan interactions with P-gp were studied using the IPEC-J2 MDR1 cell line. Lastly, in vivo neuropharmacokinetic assessment of the unbound brain-to-plasma disposition of eletriptan was conducted in wild type and mdr1a/1b knockout mice. RESULTS We demonstrated that most triptans were able to inhibit uptake of the H+/OC antiporter substrate, pyrilamine, with eletriptan emerging as the strongest inhibitor. Eletriptan, almotriptan, and sumatriptan exhibited a pH-dependent uptake into hCMEC/D3 cells. Eletriptan demonstrated saturable uptake kinetics with an apparent Km of 89 ± 38 µM and a Jmax of 2.2 ± 0.7 nmol·min-1·mg protein-1 (n = 3). Bidirectional transport experiments across IPEC-J2 MDR1 monolayers showed that eletriptan is transported by P-gp, thus indicating that eletriptan is both a substrate of the H+/OC antiporter and P-gp. This was further confirmed in vivo, where the unbound brain-to-unbound plasma concentration ratio (Kp,uu) was 0.04 in wild type mice while the ratio rose to 1.32 in mdr1a/1b knockout mice. CONCLUSIONS We have demonstrated that the triptan family of compounds possesses affinity for the H+/OC antiporter proposing that the putative H+/OC antiporter plays a role in the BBB transport of triptans, particularly eletriptan. Our in vivo studies indicate that eletriptan is subjected to simultaneous brain uptake and efflux, possibly facilitated by the putative H+/OC antiporter and P-gp, respectively. Our findings offer novel insights into the potential central site of action involved in migraine treatment with triptans and highlight the significance of potential transporter related drug-drug interactions.
Collapse
Affiliation(s)
- Nana Svane
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | - Anne Rodenberg
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Burak Ozgür
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, Denmark
| | - Lasse Saaby
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
- Bioneer: FARMA, Bioneer A/S, Copenhagen, Denmark
| | | | - Mie Kristensen
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Peer Tfelt-Hansen
- Danish Headache Center, Department of Neurology, Rigshospitalet-Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
32
|
Jaiswal N, Kumar A. Modulators of Candida albicans Membrane Drug Transporters: A Lucrative Portfolio for the Development of Effective Antifungals. Mol Biotechnol 2024; 66:960-974. [PMID: 38206530 DOI: 10.1007/s12033-023-01017-1] [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: 07/28/2023] [Accepted: 12/01/2023] [Indexed: 01/12/2024]
Abstract
The escalating prevalence of membrane drug transporters and drug efflux pumps in pathogenic yeast like Candida albicans necessitates a comprehensive understanding of their roles in MDR. The overexpression of drug transporter families, ABC and MFS, implicated in MDR through drug efflux and poses a significant challenge in the diagnosis and treatment of fungal infection. Various mechanisms have been proposed for MDR; however, the upregulation of ABC and MFS superfamily transporters is most noticeable in MDR. The direct inhibition of these transporters seems an efficient strategy to overcome this problem. The goal of the article is to present an overview of the prospect of utilizing these modulators of C. albicans drug transports as effective antifungal molecules against MDR addressing a critical gap in the field. The review tries to address to prevent drug extrusion by modulating the expression of drug transporters of C. albicans. The review discussed the progress in identifying potent, selective, and non-toxic modulators of these transporters to develop some effective antifungals and overcome MDR. We reviewed major studies in this area and found that recent work has shifted toward the exploration of natural compounds as potential modulators to restore drug sensitivity in MDR fungal cells. The focus of this review is to survey and interpret current research information on modulators of C. albicans drug transporters from natural sources emphasizing those compounds that are potent antifungal agents.
Collapse
Affiliation(s)
- Neha Jaiswal
- Department of Biotechnology, National Institute of Technology, Raipur, CG, 492010, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, CG, 492010, India.
| |
Collapse
|
33
|
Allam RM, El Kerdawy AM, Gouda AE, Ahmed KA, Abdel-Mohsen HT. Benzimidazole-oxindole hybrids as multi-kinase inhibitors targeting melanoma. Bioorg Chem 2024; 146:107243. [PMID: 38457953 DOI: 10.1016/j.bioorg.2024.107243] [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: 12/28/2023] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/10/2024]
Abstract
In the current study, a series of benzimidazole-oxindole conjugates 8a-t were designed and synthesized as type II multi-kinase inhibitors. They exhibited moderate to potent inhibitory activity against BRAFWT up to 99.61 % at 10 µM. Notably, compounds 8e, 8k, 8n and 8s demonstrated the most promising activity, with 99.44 to 99.61 % inhibition. Further evaluation revealed that 8e, 8k, 8n and 8s exhibit moderate to potent inhibitory effects on the kinases BRAFV600E, VEGFR-2, and FGFR-1. Additionally, compounds 8a-t were screened for their cytotoxicity by the NCI, and several compounds showed significant growth inhibition in diverse cancer cell lines. Compound 8e stood out with a GI50 range of 1.23 - 3.38 µM on melanoma cell lines. Encouraged by its efficacy, it was further investigated for its antitumor activity and mechanism of action, using sorafenib as a reference standard. The hybrid compound 8e exhibited potent cellular-level suppression of BRAFWT, VEGFR-2, and FGFR-1 in A375 cell line, surpassing the effects of sorafenib. In vivo studies demonstrate that 8e significantly inhibits the growth of B16F10 tumors in mice, leading to increased survival rates and histopathological tumor regression. Furthermore, 8e reduces angiogenesis markers, mRNA expression levels of VEGFR-2 and FGFR-1, and production of growth factors. It also downregulated Notch1 protein expression and decreased TGF-β1 production. Molecular docking simulations suggest that 8e binds as a promising type II kinase inhibitor in the target kinases interacting with the key regions in their kinase domain.
Collapse
Affiliation(s)
- Rasha M Allam
- Department of Pharmacology, Medical and Clinical Research Institute, National Research Centre, El-Buhouth St., Dokki, P.O. Box 12622, Cairo, Egypt
| | - Ahmed M El Kerdawy
- School of Pharmacy, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, United Kingdom; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, P.O. Box 11562, Cairo, Egypt
| | - Ahmed E Gouda
- Pharmaceutical Research Department, Nawah Scientific, Cairo, Egypt
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Heba T Abdel-Mohsen
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El-Buhouth St., Dokki, P.O. Box 12622, Cairo, Egypt.
| |
Collapse
|
34
|
He M, Zhao W, Wang P, Li W, Chen H, Yuan Z, Pan G, Gao H, Sun L, Chu J, Li L, Hu Y. Efficacy and safety of Trastuzumab Emtansine in treating human epidermal growth factor receptor 2-positive metastatic breast cancer in Chinese population: a real-world multicenter study. Front Med (Lausanne) 2024; 11:1383279. [PMID: 38741766 PMCID: PMC11089149 DOI: 10.3389/fmed.2024.1383279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/04/2024] [Indexed: 05/16/2024] Open
Abstract
Background Trastuzumab emtansine (T-DM1) has been approved worldwide for treating metastatic breast cancer (mBC) in patients who have received first-line therapy, shown disease progression, and are human epidermal growth factor receptor 2 (HER2)-positive. T-DM1 received approval in China to treat early-stage breast cancer (BC) in 2020 and for mBC in 2021. In March 2023, T-DM1 was included in medical insurance coverage, significantly expanding the eligible population. Materials and methods This post-marketing observational study aimed to assess the safety and effectiveness of T-DM1 in real-world clinical practice in China. This study enrolled 31 individuals with HER2-positive early-stage BC and 70 individuals with HER2-positive advanced BC from 8 study centers in Shandong Province, China. The T-DM1 dosage was 3.6 mg/kg injected intravenously every 3 weeks until the disease advanced or the drug toxicity became uncontrollable, whichever occurred earlier. Additionally, efficacy and safety information on T-DM1 were collected. Results During the 7-month follow-up period, no recurrence or metastases were observed in patients who had early-stage BC. The disease control rate was 31.43% (22/70) in patients with advanced BC. The most common adverse effect of T-DM1 was thrombocytopenia, with an incidence of 69.31% (70/101), and the probability of Grade ≥ 3 thrombocytopenia was 11.88% (12/101). Conclusion This real-world study demonstrated that T-DM1 had good efficacy and was well tolerated by both HER2-positive early-stage BC and mBC patients.
Collapse
Affiliation(s)
- Miao He
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
- Department of Medical Oncology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wen Zhao
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Peng Wang
- Department of Medical Oncology, Qingdao Shibei Changqing Hospital, Qingdao, Shandong Province, China
| | - Wenhuan Li
- Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Hanhan Chen
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zonghuai Yuan
- Department of General Surgery, People’s Hospital of Rizhao, Rizhao, Shandong Province, China
| | - Guangye Pan
- Department of General Surgery, People’s Hospital of Rizhao, Rizhao, Shandong Province, China
| | - Hong Gao
- Department of Breast and Thyroid Surgery, Rizhao Traditional Chinese Medical Hospital, Rizhao, Shandong Province, China
| | - Lijun Sun
- Department of Breast and Thyroid Surgery, People’s Hospital of Juxian, Rizhao, Shandong Province, China
| | - Jiahui Chu
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Li Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Yu Hu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
35
|
Qian W, Lu J, Gao C, Liu Q, Li Y, Zeng Q, Zhang J, Wang T, Chen S. Deciphering antifungal and antibiofilm mechanisms of isobavachalcone against Cryptococcus neoformans through RNA-seq and functional analyses. Microb Cell Fact 2024; 23:107. [PMID: 38609931 PMCID: PMC11015616 DOI: 10.1186/s12934-024-02369-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
Cryptococcus neoformans has been designated as critical fungal pathogens by the World Health Organization, mainly due to limited treatment options and the prevalence of antifungal resistance. Consequently, the utilization of novel antifungal agents is crucial for the effective treatment of C. neoformans infections. This study exposed that the minimum inhibitory concentration (MIC) of isobavachalcone (IBC) against C. neoformans H99 was 8 µg/mL, and IBC dispersed 48-h mature biofilms by affecting cell viability at 16 µg/mL. The antifungal efficacy of IBC was further validated through microscopic observations using specific dyes and in vitro assays, which confirmed the disruption of cell wall/membrane integrity. RNA-Seq analysis was employed to decipher the effect of IBC on the C. neoformans H99 transcriptomic profiles. Real-time quantitative reverse transcription PCR (RT-qPCR) analysis was performed to validate the transcriptomic data and identify the differentially expressed genes. The results showed that IBC exhibited various mechanisms to impede the growth, biofilm formation, and virulence of C. neoformans H99 by modulating multiple dysregulated pathways related to cell wall/membrane, drug resistance, apoptosis, and mitochondrial homeostasis. The transcriptomic findings were corroborated by the antioxidant analyses, antifungal drug sensitivity, molecular docking, capsule, and melanin assays. In vivo antifungal activity analysis demonstrated that IBC extended the lifespan of C. neoformans-infected Caenorhabditis elegans. Overall, the current study unveiled that IBC targeted multiple pathways simultaneously to inhibit growth significantly, biofilm formation, and virulence, as well as to disperse mature biofilms of C. neoformans H99 and induce cell death.
Collapse
Affiliation(s)
- Weidong Qian
- School of Biological and Pharmaceutical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China.
| | - Jiaxing Lu
- School of Biological and Pharmaceutical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Chang Gao
- School of Biological and Pharmaceutical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Qiming Liu
- School of Biological and Pharmaceutical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yongdong Li
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, 315010, P. R. China
| | - Qiao Zeng
- School of Biological and Pharmaceutical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Ting Wang
- School of Biological and Pharmaceutical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Si Chen
- Department of Immunology, Shenzhen University Medical School, Shenzhen, 518060, China.
| |
Collapse
|
36
|
Tilocca B, Greco V, Piras C, Ceniti C, Paonessa M, Musella V, Bava R, Palma E, Morittu VM, Spina AA, Castagna F, Urbani A, Britti D, Roncada P. The Bee Gut Microbiota: Bridging Infective Agents Potential in the One Health Context. Int J Mol Sci 2024; 25:3739. [PMID: 38612550 PMCID: PMC11012054 DOI: 10.3390/ijms25073739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The bee gut microbiota plays an important role in the services the bees pay to the environment, humans and animals. Alongside, gut-associated microorganisms are vehiculated between apparently remote habitats, promoting microbial heterogeneity of the visited microcosms and the transfer of the microbial genetic elements. To date, no metaproteomics studies dealing with the functional bee microbiota are available. Here, we employ a metaproteomics approach to explore a fraction of the bacterial, fungal, and unicellular parasites inhabiting the bee gut. The bacterial community portrays a dynamic composition, accounting for specimens of human and animal concern. Their functional features highlight the vehiculation of virulence and antimicrobial resistance traits. The fungal and unicellular parasite fractions include environment- and animal-related specimens, whose metabolic activities support the spatial spreading of functional features. Host proteome depicts the major bee physiological activities, supporting the metaproteomics strategy for the simultaneous study of multiple microbial specimens and their host-crosstalks. Altogether, the present study provides a better definition of the structure and function of the bee gut microbiota, highlighting its impact in a variety of strategies aimed at improving/overcoming several current hot topic issues such as antimicrobial resistance, environmental pollution and the promotion of environmental health.
Collapse
Affiliation(s)
- Bruno Tilocca
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Viviana Greco
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Hearth, 00168 Rome, Italy; (V.G.); (A.U.)
- Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Diagnostic and Laboratory Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Cristian Piras
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Carlotta Ceniti
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Mariachiara Paonessa
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Vincenzo Musella
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Roberto Bava
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Ernesto Palma
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Valeria Maria Morittu
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Anna Antonella Spina
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Fabio Castagna
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Hearth, 00168 Rome, Italy; (V.G.); (A.U.)
- Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Diagnostic and Laboratory Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Domenico Britti
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| | - Paola Roncada
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (C.P.); (C.C.); (M.P.); (V.M.); (R.B.); (E.P.); (V.M.M.); (A.A.S.); (F.C.); (D.B.)
| |
Collapse
|
37
|
Huang Z, Zhu W, Bai Y, Bai X, Zhang H. Non-ribosomal peptide synthetase (NRPS)-encoding products and their biosynthetic logics in Fusarium. Microb Cell Fact 2024; 23:93. [PMID: 38539193 PMCID: PMC10967133 DOI: 10.1186/s12934-024-02378-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/21/2024] [Indexed: 11/11/2024] Open
Abstract
Fungal non-ribosomal peptide synthetase (NRPS)-encoding products play a paramount role in new drug discovery. Fusarium, one of the most common filamentous fungi, is well-known for its biosynthetic potential of NRPS-type compounds with diverse structural motifs and various biological properties. With the continuous improvement and extensive application of bioinformatic tools (e.g., anti-SMASH, NCBI, UniProt), more and more biosynthetic gene clusters (BGCs) of secondary metabolites (SMs) have been identified in Fusarium strains. However, the biosynthetic logics of these SMs have not yet been well investigated till now. With the aim to increase our knowledge of the biosynthetic logics of NPRS-encoding products in Fusarium, this review firstly provides an overview of research advances in elucidating their biosynthetic pathways.
Collapse
Affiliation(s)
- Ziwei Huang
- School of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Wangjie Zhu
- School of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yifan Bai
- School of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xuelian Bai
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Huawei Zhang
- School of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
38
|
Karamali N, Daraei A, Rostamlou A, Mahdavi R, Akbari Jonoush Z, Ghadiri N, Mahmoudi Z, Mardi A, Javidan M, Sohrabi S, Baradaran B. Decoding contextual crosstalk: revealing distinct interactions between non-coding RNAs and unfolded protein response in breast cancer. Cancer Cell Int 2024; 24:104. [PMID: 38468244 PMCID: PMC10926595 DOI: 10.1186/s12935-024-03296-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/06/2024] [Indexed: 03/13/2024] Open
Abstract
Breast cancer is significantly influenced by endoplasmic reticulum (ER) stress, impacting both its initiation and progression. When cells experience an accumulation of misfolded or unfolded proteins, they activate the unfolded protein response (UPR) to restore cellular balance. In breast cancer, the UPR is frequently triggered due to challenging conditions within tumors. The UPR has a dual impact on breast cancer. On one hand, it can contribute to tumor growth by enhancing cell survival and resistance to programmed cell death in unfavorable environments. On the other hand, prolonged and severe ER stress can trigger cell death mechanisms, limiting tumor progression. Furthermore, ER stress has been linked to the regulation of non-coding RNAs (ncRNAs) in breast cancer cells. These ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play essential roles in cancer development by influencing gene expression and cellular processes. An improved understanding of how ER stress and ncRNAs interact in breast cancer can potentially lead to new treatment approaches. Modifying specific ncRNAs involved in the ER stress response might interfere with cancer cell survival and induce cell death. Additionally, focusing on UPR-associated proteins that interact with ncRNAs could offer novel therapeutic possibilities. Therefore, this review provides a concise overview of the interconnection between ER stress and ncRNAs in breast cancer, elucidating the nuanced effects of the UPR on cell fate and emphasizing the regulatory roles of ncRNAs in breast cancer progression.
Collapse
Affiliation(s)
- Negin Karamali
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arshia Daraei
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Arman Rostamlou
- Department of Medical Biology, School of Medicine, University of EGE, Bornova, Izmir, Turkey
| | - Roya Mahdavi
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Akbari Jonoush
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nooshin Ghadiri
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Mahmoudi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Mardi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Javidan
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sepideh Sohrabi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
39
|
Chen DQ, Xie Y, Cao LQ, Fleishman JS, Chen Y, Wu T, Yang DH. The role of ABCC10/MRP7 in anti-cancer drug resistance and beyond. Drug Resist Updat 2024; 73:101062. [PMID: 38330827 DOI: 10.1016/j.drup.2024.101062] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
Multidrug resistance protein 7 (MRP7), also known as ATP-binding cassette (ABC) transporter subfamily C10 (ABCC10), is an ABC transporter that was first identified in 2001. ABCC10/MRP7 is a 171 kDa protein located on the basolateral membrane of cells. ABCC10/MRP7 consists of three transmembrane domains and two nucleotide binding domains. It mediates multidrug resistance of tumor cells to a variety of anticancer drugs by increasing drug efflux and results in reducing intracellular drug accumulation. The transport substrates of ABCC10/MRP7 include antineoplastic drugs such as taxanes, vinca alkaloids, and epothilone B, as well as endobiotics such as leukotriene C4 (LTC4) and estradiol 17 β-D-glucuronide. A variety of ABCC10/MRP7 inhibitors, including cepharanthine, imatinib, erlotinib, tariquidar, and sildenafil, can reverse ABCC10/MRP7-mediated MDR. Additionally, the presence or absence of ABCC10/MRP7 is also closely related to renal tubular dysfunction, obesity, and other diseases. In this review, we discuss: 1) Structure and functions of ABCC10/MRP7; 2) Known substrates and inhibitors of ABCC10/MRP7 and their potential therapeutic applications in cancer; and 3) Role of ABCC10/MRP7 in non-cancerous diseases.
Collapse
Affiliation(s)
- Da-Qian Chen
- Department of Medical Oncology, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong 518100, China
| | - Yuhao Xie
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Lu-Qi Cao
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Institute for Biotechnology, St. John's University, Queens, NY 11439, USA
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yang Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Tiesong Wu
- Department of Pharmacy, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong 518100, China.
| | - Dong-Hua Yang
- Department of Medical Oncology, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong 518100, China; New York College of Traditional Chinese Medicine, Mineola, NY 11501, USA.
| |
Collapse
|
40
|
Boussaha S, Lassed S, Abdelwahab AB, Krid A, Altun M, Chalard PP, Chalchat PJC, Figueredo G, Zama PD, Demirtas PI, Benayache PS, Benayache PF. Chemical Characterization, DNA-Damage Protection, Antiproliferative Activity and in Silico Studies of the Essential Oils from Perralderia coronopifolia Coss. Chem Biodivers 2024; 21:e202301535. [PMID: 38010960 DOI: 10.1002/cbdv.202301535] [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: 09/29/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 11/29/2023]
Abstract
In this study, for the first time, we analyzed the chemical composition of essential oils (EOs) steam-distilled from the flowers and leaves of Perralderia coronopifolia by GC-FID/MS. The objective was to explore new anticancer and antioxidant bioactive substances and understand their mechanisms of action through the use of plant-derived natural products. The major chemical components characterizing the EOs were cis-chrysanthenyl acetate 1, 6-oxocyclonerolidol 2, cis-8-acetoxychrysanthenyl acetate 3, and 6α-hydroxycyclonerolidol 4, respectively. Furthermore, the EOs inhibited cell proliferation in HeLa (human cervix carcinoma) and PC3 (human prostate cancer) cells and protected plasmid DNA from oxidative damage caused by UV-photolyzed H2 O2 . Employing a molecular docking study, we elucidated the main compounds' inhibition mechanisms. Consequently, the antitumor activity could be related to the inhibitory property of compound 3 against CDC25B phosphatase. The evaluation of ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties and the density functional theory (DFT) calculations of the major compounds, especially compound 3, offer potential insights for designing and developing new cancer drug candidates. In conclusion, our study provides a framework for future research and development in the field by establishing a scientific foundation for the use of Perralderia coronopifolia essential oils as a prospective source of antioxidant and anticancer agents.
Collapse
Affiliation(s)
- Sara Boussaha
- Unité de Recherche: Valorisation des Ressources Naturelles, Molécules Bioactives et Analyses Physicochimiques et Biologiques, Université Frères Mentouri, Constantine 1. Route d'Aïn El Bey, 25017, Constantine, Algérie
- Higher National School of Biotechnology Taoufik KHAZNADAR, nouveau Pôle universitaire Ali Mendili, BP. E66, Constantine, 25100, Algeria
| | - Somia Lassed
- Département de Microbiologie et Biochimie, Université Mostefa Benboulaid, Batna-2, 05078, Batna, Algérie
| | - Ahmed B Abdelwahab
- Temisis Therapeutics, 19 avenue de la Forêt de Haye, 54500, Vandœuvre-lès-Nancy, France
| | - Adel Krid
- Laboratoire de Physique Mathématique et Subatomique LPMS, Département de Chimie, Université des Frères Mentouri, 25017, Constantine, Algeria
- Pharmaceutical Sciences Research Center (CRSP), Ali Mendjli, Constantine, 25000, Algeria
| | - Muhammed Altun
- Plant research laboratory, Chemistry Department, Cankiri Karatekin University, Ballica Campus, 18100, Cankiri, Turkey
| | - Pr Pierre Chalard
- Université Clermont Auvergne, CNRS SIGMA Clermont ICC, F-63000, Clermont Ferrand, France
| | - Pr Jean Claude Chalchat
- Association de Valorisation des Huiles Essentielles et des Arômes (AVAHEA), La Laye 7, 63500, Saint Babel, France
| | - Gilles Figueredo
- Laboratoire d'Analyses des Extraits Végétaux et des Arômes (LEXVA Analytique), 460 Rue du Montant, 63110, Beaumont, France
| | - Pr Djamila Zama
- Unité de Recherche: Valorisation des Ressources Naturelles, Molécules Bioactives et Analyses Physicochimiques et Biologiques, Université Frères Mentouri, Constantine 1. Route d'Aïn El Bey, 25017, Constantine, Algérie
| | - Pr Ibrahim Demirtas
- Plant research laboratory, Chemistry Department, Cankiri Karatekin University, Ballica Campus, 18100, Cankiri, Turkey
| | - Pr Samir Benayache
- Unité de Recherche: Valorisation des Ressources Naturelles, Molécules Bioactives et Analyses Physicochimiques et Biologiques, Université Frères Mentouri, Constantine 1. Route d'Aïn El Bey, 25017, Constantine, Algérie
| | - Pr Fadila Benayache
- Unité de Recherche: Valorisation des Ressources Naturelles, Molécules Bioactives et Analyses Physicochimiques et Biologiques, Université Frères Mentouri, Constantine 1. Route d'Aïn El Bey, 25017, Constantine, Algérie
| |
Collapse
|
41
|
Shobana R, Thahirunnisa JH, Sivaprakash S, Amali AJ, Solomon AP, Suresh D. Effect of palladium(II) complexes on NorA efflux pump inhibition and resensitization of fluoroquinolone-resistant Staphylococcus aureus: in vitro and in silico approach. Front Cell Infect Microbiol 2024; 13:1340135. [PMID: 38292858 PMCID: PMC10825952 DOI: 10.3389/fcimb.2023.1340135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/20/2023] [Indexed: 02/01/2024] Open
Abstract
Staphylococcus aureus leads to diverse infections, and their treatment relies on the use of antibiotics. Nevertheless, the rise of antibiotic resistance poses an escalating challenge and various mechanisms contribute to antibiotic resistance, including modifications to drug targets, enzymatic deactivation of drugs, and increased efflux of antibiotics. Hence, the quest for innovative antimicrobial solutions has intensified in the face of escalating antibiotic resistance and the looming threat of superbugs. The NorA protein of S. aureus, classified as an efflux pump within the major facilitator superfamily, when overexpressed, extrudes various substances, including fluoroquinolones (such as ciprofloxacin) and quaternary ammonium. Addressing this, the unexplored realm of inorganic and organometallic compounds in medicinal chemistry holds promise. Notably, the study focused on investigating two different series of palladium-based metal complexes consisting of QSL_PA and QSL_PB ligands to identify a potent NorA efflux pump inhibitor that can restore the susceptibility to fluoroquinolone antibiotics. QSL_Pd5A was identified as a potent efflux pump inhibitor from the real-time efflux assay. QSL_Pd5A also resensitized SA1199B to ciprofloxacin at a low concentration of 0.125 µg/mL without elucidating cytotoxicity on the NRK-62E cell line. The in vitro findings were substantiated by docking results, indicating favorable interactions between QSL_Pd5A and the NorA efflux pump.
Collapse
Affiliation(s)
- Rajaramon Shobana
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Jaffer Hussain Thahirunnisa
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Selvam Sivaprakash
- Organometallics and Catalysis Laboratory, Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Arlin Jose Amali
- Organometallics and Catalysis Laboratory, Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Devarajan Suresh
- Organometallics and Catalysis Laboratory, Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| |
Collapse
|
42
|
Xue WH, Liu KL, Zhang TJ, Dong G, Wang JH, Wang J, Guo S, Hu J, Zhang QY, Li XY, Meng FH. Discovery of (quinazolin-6-yl)benzamide derivatives containing a 6,7-dimethoxy-1,2,3,4-tetrahydroisoquinoline moiety as potent reversal agents against P-glycoprotein-mediated multidrug resistance. Eur J Med Chem 2024; 264:116039. [PMID: 38103540 DOI: 10.1016/j.ejmech.2023.116039] [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: 07/12/2023] [Revised: 08/28/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
P-glycoprotein (P-gp) is an important factor leading to multidrug resistance (MDR) in cancer treatment. The co-administration of anticancer drugs and P-gp inhibitors has been a treatment strategy to overcome MDR. In recent years, tyrosine kinase inhibitor Lapatinib has been reported to reverse MDR through directly interacting with ABC transporters. In this work, a series of P-gp inhibitors (1-26) was designed and synthesized by integrating the quinazoline core of Lapatinib into the molecule framework of the third-generation P-gp inhibitor Tariquidar. Among them, compound 14 exhibited better MDR reversal activity than Tariquidar. The docking results showed compound 14 displayed the L-shaped molecular conformation. Importantly, compound 14 increased the accumulation of Adriamycin (ADM) and rhodamine 123 (Rh123) in MCF7/ADM cells. Besides, compound 14 significantly increased ADM-induced apoptosis and inhibited the proliferation, migration and invasion of MCF7/ADM cells. It was also demonstrated that compound 14 significantly inhibited the growth of MCF7/ADM xenograft tumors by increasing the sensitivity of ADM. In summary, compound 14 has the potential to overcome MDR caused by P-gp.
Collapse
Affiliation(s)
- Wen-Han Xue
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Kai-Li Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Ting-Jian Zhang
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Gang Dong
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Jia-Hui Wang
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Jing Wang
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Shuai Guo
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Jie Hu
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Qing-Yu Zhang
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China
| | - Xin-Yang Li
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, Shenyang, 110122, PR China.
| |
Collapse
|
43
|
Lim YJ, Kim HS, Bae S, So KA, Kim TJ, Lee JH. Pan-EGFR Inhibitor Dacomitinib Resensitizes Paclitaxel and Induces Apoptosis via Elevating Intracellular ROS Levels in Ovarian Cancer SKOV3-TR Cells. Molecules 2024; 29:274. [PMID: 38202856 PMCID: PMC10780346 DOI: 10.3390/molecules29010274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Paclitaxel is still used as a standard first-line treatment for ovarian cancer. Although paclitaxel is effective for many types of cancer, the emergence of chemoresistant cells represents a major challenge in chemotherapy. Our study aimed to analyze the cellular mechanism of dacomitinib, a pan-epidermal growth factor receptor (EGFR) inhibitor, which resensitized paclitaxel and induced cell cytotoxicity in paclitaxel-resistant ovarian cancer SKOV3-TR cells. We investigated the significant reduction in cell viability cotreated with dacomitinib and paclitaxel by WST-1 assay and flow cytometry analysis. Dacomitinib inhibited EGFR family proteins, including EGFR and HER2, as well as its downstream signaling proteins, including AKT, STAT3, ERK, and p38. In addition, dacomitinib inhibited the phosphorylation of Bad, and combination treatment with paclitaxel effectively suppressed the expression of Mcl-1. A 2'-7'-dichlorodihydrofluorescein diacetate (DCFH-DA) assay revealed a substantial elevation in cellular reactive oxygen species (ROS) levels in SKOV3-TR cells cotreated with dacomitinib and paclitaxel, which subsequently mediated cell cytotoxicity. Additionally, we confirmed that dacomitinib inhibits chemoresistance in paclitaxel-resistant ovarian cancer HeyA8-MDR cells. Collectively, our research indicated that dacomitinib effectively resensitized paclitaxel in SKOV3-TR cells by inhibiting EGFR signaling and elevating intracellular ROS levels.
Collapse
Affiliation(s)
- Ye Jin Lim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Hee Su Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Seunghee Bae
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Kyeong A So
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Jae Ho Lee
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| |
Collapse
|
44
|
Pandey P, Garg A, Singh V, Rai G, Mishra N. Clinical Trials and Future Prospects of Autophagy and ROS in Cancer. CANCER DRUG DISCOVERY AND DEVELOPMENT 2024:337-369. [DOI: 10.1007/978-3-031-66421-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
45
|
Ahmadpour ST, Orre C, Bertevello PS, Mirebeau-Prunier D, Dumas JF, Desquiret-Dumas V. Breast Cancer Chemoresistance: Insights into the Regulatory Role of lncRNA. Int J Mol Sci 2023; 24:15897. [PMID: 37958880 PMCID: PMC10650504 DOI: 10.3390/ijms242115897] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are a subclass of noncoding RNAs composed of more than 200 nucleotides without the ability to encode functional proteins. Given their involvement in critical cellular processes such as gene expression regulation, transcription, and translation, lncRNAs play a significant role in organism homeostasis. Breast cancer (BC) is the second most common cancer worldwide and evidence has shown a relationship between aberrant lncRNA expression and BC development. One of the main obstacles in BC control is multidrug chemoresistance, which is associated with the deregulation of multiple mechanisms such as efflux transporter activity, mitochondrial metabolism reprogramming, and epigenetic regulation as well as apoptosis and autophagy. Studies have shown the involvement of a large number of lncRNAs in the regulation of such pathways. However, the underlying mechanism is not clearly elucidated. In this review, we present the principal mechanisms associated with BC chemoresistance that can be directly or indirectly regulated by lncRNA, highlighting the importance of lncRNA in controlling BC chemoresistance. Understanding these mechanisms in deep detail may interest the clinical outcome of BC patients and could be used as therapeutic targets to overcome BC therapy resistance.
Collapse
Affiliation(s)
- Seyedeh Tayebeh Ahmadpour
- Nutrition, Croissance et Cancer, Inserm, UMR1069, Université de Tours, 37032 Tours, France; (P.S.B.); (J.-F.D.)
| | - Charlotte Orre
- Inserm U1083, UMR CNRS 6214, Angers University, 49933 Angers, France; (C.O.); (D.M.-P.)
| | - Priscila Silvana Bertevello
- Nutrition, Croissance et Cancer, Inserm, UMR1069, Université de Tours, 37032 Tours, France; (P.S.B.); (J.-F.D.)
| | | | - Jean-François Dumas
- Nutrition, Croissance et Cancer, Inserm, UMR1069, Université de Tours, 37032 Tours, France; (P.S.B.); (J.-F.D.)
| | | |
Collapse
|
46
|
Mossel P, Arif WM, De Souza GS, Varela LG, van der Weijden CWJ, Boersma HH, Willemsen ATM, Boellaard R, Elsinga PH, Borra RJH, Dierckx RAJO, Lammertsma AA, Bartels AL, Luurtsema G. Quantification of P-glycoprotein function at the human blood-brain barrier using [ 18F]MC225 and PET. Eur J Nucl Med Mol Imaging 2023; 50:3917-3927. [PMID: 37552369 PMCID: PMC10611838 DOI: 10.1007/s00259-023-06363-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION P-glycoprotein (P-gp) is one of the most studied efflux transporters at the blood-brain barrier. It plays an important role in brain homeostasis by protecting the brain from a variety of endogenous and exogeneous substances. Changes in P-gp function are associated both with the onset of neuropsychiatric diseases, including Alzheimer's disease and Parkinson's disease, and with drug-resistance, for example in treatment-resistant depression. The most widely used approach to measure P-gp function in vivo is (R)-[11C]verapamil PET. (R)-[11C]verapamil is, however, an avid P-gp substrate, which complicates the use of this tracer to measure an increase in P-gp function as its baseline uptake is already very low. [18F]MC225 was developed to measure both increases and decreases in P-gp function. AIM The aim of this study was (1) to identify the pharmacokinetic model that best describes [18F]MC225 kinetics in the human brain and (2) to determine test-retest variability. METHODS Five (2 male, 3 female) of fourteen healthy subjects (8 male, 6 female, age 67 ± 5 years) were scanned twice (injected dose 201 ± 47 MBq) with a minimum interval of 2 weeks between scans. Each scanning session consisted of a 60-min dynamic [18F]MC225 scan with continuous arterial sampling. Whole brain grey matter data were fitted to a single tissue compartment model, and to reversible and irreversible two tissue-compartment models to obtain various outcome parameters (in particular the volume of distribution (VT), Ki, and the rate constants K1 and k2). In addition, a reversible two-tissue compartment model with fixed k3/k4 was included. The preferred model was selected based on the weighted Akaike Information Criterion (AIC) score. Test-retest variability (TRTV) was determined to assess reproducibility. RESULTS Sixty minutes post-injection, the parent fraction was 63.8 ± 4.0%. The reversible two tissue compartment model corrected for plasma metabolites with an estimated blood volume (VB) showed the highest AIC weight score of 34.3 ± 17.6%. The TRVT of the VT for [18F]MC225 PET scans was 28.3 ± 20.4% for the whole brain grey matter region using this preferred model. CONCLUSION [18F]MC225 VT, derived using a reversible two-tissue compartment model, is the preferred parameter to describe P-gp function in the human BBB. This outcome parameter has an average test-retest variability of 28%. TRIAL REGISTRATION EudraCT 2020-001564-28 . Registered 25 May 2020.
Collapse
Affiliation(s)
- Pascalle Mossel
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wejdan M Arif
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- College of Applied Medical Science, Department of Radiological Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Giordana Salvi De Souza
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lara Garcia Varela
- Molecular Imaging Biomarkers Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), 15706, Santiago de Compostela, Spain
- Nuclear Medicine Department and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706, Santiago de Compostela, Spain
| | - Chris W J van der Weijden
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hendrikus H Boersma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Antoon T M Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC, Location VUmc, Amsterdam, The Netherlands
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronald J H Borra
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna L Bartels
- Department of Neurology, Ommelander Ziekenhuis Groep, Scheemda, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
47
|
Cho Y, Hwang JW, Park NJ, Moon J, Ali KH, Seo YH, Kim IS, Kim SN, Kim YK. SPC-180002, a SIRT1/3 dual inhibitor, impairs mitochondrial function and redox homeostasis and represents an antitumor activity. Free Radic Biol Med 2023; 208:73-87. [PMID: 37536458 DOI: 10.1016/j.freeradbiomed.2023.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/12/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Since sirtuins (SIRTs) are closely associated with reactive oxygen species (ROS) and antioxidant system, the development of their selective inhibitors is drawing attention for understanding of cellular redox homeostasis. Here, we describe the pharmacological properties of SPC-180002, which incorporates a methyl methacrylate group as a key pharmacophore, along with its comprehensive molecular mechanism as a novel dual inhibitor of SIRT1/3. The dual inhibition of SIRT1/3 by SPC-180002 disturbs redox homeostasis via ROS generation, which leads to an increase in both p21 protein stability and mitochondrial dysfunction. Increased p21 interacts with and inhibits CDK, thereby interfering with cell cycle progression. SPC-180002 leads to mitochondrial dysfunction by inhibiting mitophagy, which is accompanied by a reduction in oxygen consumption rate. Consequently, SPC-180002 strongly suppresses the proliferation of cancer cells and exerts anticancer effect in vivo. Taken together, the novel SIRT1/3 dual inhibitor, SPC-180002, impairs mitochondrial function and redox homeostasis, thereby strongly inhibiting cell cycle progression and cancer cell growth.
Collapse
Affiliation(s)
- Yena Cho
- Muscle Physiome Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jee Won Hwang
- Muscle Physiome Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - No-June Park
- Natural Product Research Institute, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea; Division of Bio-Medical Science and Technology, University of Science and Technology KIST School, Seoul, 02792, Republic of Korea
| | - Junghyea Moon
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Khan Hashim Ali
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Young Ho Seo
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Su-Nam Kim
- Natural Product Research Institute, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea; Division of Bio-Medical Science and Technology, University of Science and Technology KIST School, Seoul, 02792, Republic of Korea.
| | - Yong Kee Kim
- Muscle Physiome Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
48
|
Marjamaa A, Gibbs B, Kotrba C, Masamha CP. The role and impact of alternative polyadenylation and miRNA regulation on the expression of the multidrug resistance-associated protein 1 (MRP-1/ABCC1) in epithelial ovarian cancer. Sci Rep 2023; 13:17476. [PMID: 37838788 PMCID: PMC10576765 DOI: 10.1038/s41598-023-44548-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023] Open
Abstract
The ATP-binding cassette transporter (ABCC1) is associated with poor survival and chemotherapy drug resistance in high grade serous ovarian cancer (HGSOC). The mechanisms driving ABCC1 expression are poorly understood. Alternative polyadenylation (APA) can give rise to ABCC1 mRNAs which differ only in the length of their 3'untranslated regions (3'UTRs) in a process known as 3'UTR-APA. Like other ABC transporters, shortening of the 3'UTR of ABCC1 through 3'UTR-APA would eliminate microRNA binding sites found within the longer 3'UTRs, hence eliminating miRNA regulation and altering gene expression. We found that the HGSOC cell lines Caov-3 and Ovcar-3 express higher levels of ABCC1 protein than normal cells. APA of ABCC1 occurs in all three cell lines resulting in mRNAs with both short and long 3'UTRs. In Ovcar-3, mRNAs with shorter 3'UTRs dominate resulting in a six-fold increase in protein expression. We were able to show that miR-185-5p and miR-326 both target the ABCC1 3'UTR. Hence, 3'UTR-APA should be considered as an important regulator of ABCC1 expression in HGSOC. Both HGSOC cell lines are cisplatin resistant, and we used erastin to induce ferroptosis, an alternative form of cell death. We showed that we could induce ferroptosis and sensitize the cisplatin resistant cells to cisplatin by using erastin. Knocking down ABCC1 resulted in decreased cell viability, but did not contribute to erastin induced ferroptosis.
Collapse
Affiliation(s)
- Audrey Marjamaa
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN, 46208, USA
| | - Bettine Gibbs
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, IN, 46208, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Chloe Kotrba
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, IN, 46208, USA
| | | |
Collapse
|
49
|
Mattioli R, Ilari A, Colotti B, Mosca L, Fazi F, Colotti G. Doxorubicin and other anthracyclines in cancers: Activity, chemoresistance and its overcoming. Mol Aspects Med 2023; 93:101205. [PMID: 37515939 DOI: 10.1016/j.mam.2023.101205] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Anthracyclines have been important and effective treatments against a number of cancers since their discovery. However, their use in therapy has been complicated by severe side effects and toxicity that occur during or after treatment, including cardiotoxicity. The mode of action of anthracyclines is complex, with several mechanisms proposed. It is possible that their high toxicity is due to the large set of processes involved in anthracycline action. The development of resistance is a major barrier to successful treatment when using anthracyclines. This resistance is based on a series of mechanisms that have been studied and addressed in recent years. This work provides an overview of the anthracyclines used in cancer therapy. It discusses their mechanisms of activity, toxicity, and chemoresistance, as well as the approaches used to improve their activity, decrease their toxicity, and overcome resistance.
Collapse
Affiliation(s)
- Roberto Mattioli
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy
| | - Beatrice Colotti
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Luciana Mosca
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
| |
Collapse
|
50
|
Rao ZZ, Tang ZW, Wen J. Advances in drug resistance of triple negative breast cancer caused by pregnane X receptor. World J Clin Oncol 2023; 14:335-342. [PMID: 37771631 PMCID: PMC10523191 DOI: 10.5306/wjco.v14.i9.335] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/17/2023] [Accepted: 08/29/2023] [Indexed: 09/20/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide. Triple-negative breast cancer (TNBC), refers breast cancer negative for estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2, characterized by high drug resistance, high metastasis and high recurrence, treatment of which is a difficult problem in the clinical treatment of breast cancer. In order to better treat TNBC clinically, it is a very urgent task to explore the mechanism of TNBC resistance in basic breast cancer research. Pregnane X receptor (PXR) is a nuclear receptor whose main biological function is to participate in the metabolism, transport and clearance of allobiological agents in PXR. PXR plays an important role in drug metabolism and clearance, and PXR is highly expressed in tumor tissues of TNBC patients, which is related to the prognosis of breast cancer patients. This reviews synthesized the important role of PXR in the process of high drug resistance to TNBC chemotherapeutic drugs and related research progress.
Collapse
Affiliation(s)
- Zhou-Zhou Rao
- Department of Physiology, Hunan Normal University School of Medicine, Changsha 410003, Hunan Province, China
| | - Zhong-Wen Tang
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| |
Collapse
|