1
|
Edvall C, Kale N, Tani S, Ambhore S, Hossain R, Ozoude C, Van Horsen K, Mohammad J, Tuvin DM, Kalathingal S, Loganathan J, Choi Y, Sathish V, Brown J, Mallik S. Hypoxia-Responsive Polymersomes for Stemness Reduction in Patient-Derived Solid Tumor Spheroids. ACS APPLIED BIO MATERIALS 2025; 8:2916-2926. [PMID: 40056142 DOI: 10.1021/acsabm.4c01735] [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] [Indexed: 03/10/2025]
Abstract
Aggressive solid tumors are associated with rapid growth, early hypoxia, a lack of targeted therapies, and a poor prognosis. The hypoxic niches within the rapidly growing solid tumors give rise to a stem-cell-like phenotype with higher metastasis and drug resistance. To overcome the drug resistance of these regions, we used hypoxia-responsive polymersomes with an encapsulated anticancer drug (doxorubicin, Dox) and a stemness modulator (all-trans retinoic acid, ATRA). Reductase enzymes overexpressed in hypoxia reduce the azobenzene linker of the polymers, disrupt the bilayer structure of the polymersomes, and release the encapsulated drugs. We used triple-negative breast cancer (TNBC) as a representative of aggressive and hypoxic solid tumors. We observed that ATRA synergistically enhanced the efficacy of Dox in killing cancer cells. A synergistic combination of the two drug-encapsulated polymersomes reduced the volumes of patient-derived TNBC spheroids by 90%. In contrast, Dox alone decreased the spheroid volumes by 70% and encapsulated ATRA by 19%. Mechanistic studies revealed that ATRA inhibited efflux pumps, leading to a higher concentration of doxorubicin within TNBC cells. In addition, the combination of encapsulated Dox and ATRA significantly decreased stemness expression of the TNBC cells in hypoxia compared to that of Dox alone.
Collapse
Affiliation(s)
- Connor Edvall
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Narendra Kale
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Sakurako Tani
- Department of Physics, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Shubhashri Ambhore
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Rayat Hossain
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Chukwuebuka Ozoude
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Karl Van Horsen
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jiyan Mohammad
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Daniel M Tuvin
- Sanford Broadway Clinic,801 Broadway N, Fargo, North Dakota 58102, United States
| | - Santo Kalathingal
- Agathos Biologics,4837 Amber Valley Pkwy Suite 12, Fargo, North Dakota 58104, United States
| | - Jagadish Loganathan
- Agathos Biologics,4837 Amber Valley Pkwy Suite 12, Fargo, North Dakota 58104, United States
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - James Brown
- Agathos Biologics,4837 Amber Valley Pkwy Suite 12, Fargo, North Dakota 58104, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, South Dakota 57007, United States
| |
Collapse
|
2
|
Traber GM, Tu MJ, Guan S, Batra N, Yu AM. Bioengineered miR-7-5p modulates non-small cell lung cancer cell metabolism to improve therapy. Mol Pharmacol 2025; 107:100006. [PMID: 39919164 DOI: 10.1016/j.molpha.2024.100006] [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: 09/09/2024] [Accepted: 11/14/2024] [Indexed: 02/09/2025] Open
Abstract
Reintroduction of tumor-suppressive microRNA-7-5p (miR-7) that is depleted in non-small cell lung cancer (NSCLC) represents a new therapeutic approach, whereas previous studies mainly used miR-7 mimics chemoengineered in vitro. Here we aim to establish the pharmacological actions and therapeutic potential of novel bioengineered RNA bearing a payload miR-7 (BioRNA/miR-7) molecule produced in vivo. First, through confocal imaging and immunoblot studies, we revealed that BioRNA/miR-7 altered NSCLC cell mitochondrial morphology accompanied by the downregulation of known target genes, epidermal growth factor receptor (EGFR), mitochondrial solute carrier family 25A37 (SLC25A37), and import inner membrane translocase subunit (TIM50). Second, through luciferase reporter and immunoblot studies, we validated mitochondrial acylglycerol kinase (AGK) as a new direct target for miR-7. Third, through real-time live-cell analyses, we revealed BioRNA/miR-7 to modulate mitochondrial respiration and glycolytic capacity. Fourth, live-cell and endpoint viability studies demonstrated that the combination of BioRNA/miR-7 with pemetrexed (PEM) elicited a strong synergistic effect to inhibit NSCLC cell growth, associated with an increased intracellular PEM accumulation, as quantified by a liquid chromatography tandem mass spectrometry method. Finally, through in vivo therapy study using NSCLC patient-derived xenograft mouse model, we demonstrated the efficacy and tolerability of BioRNA/miR-7 monotherapy and combination therapy with PEM to control tumor progression. Our collective works establish a role for miR-7 in NSCLC metabolism and PEM disposition and support our novel, in vivo produced BioRNA/miR-7-5p for molecular pharmacological research. Our findings further illustrate the potential of BioRNA/miR-7 plus PEM combination as a potential treatment to combat NSCLC tumor progression. SIGNIFICANCE STATEMENT: MiR-7 is a tumor-suppressive microRNA depleted in non-small cell lung cancer (NSCLC), and in vitro chemoengineered miR-7 mimics were shown to inhibit tumor growth in NSCLC cell-derived xenograft mice. Here, a novel in vivo bioengineered miR-7 molecule, namely BioRNA/miR-7, was used to effectively control target gene expression and NSCLC cell metabolism. Furthermore, BioRNA/miR-7 was demonstrated to remarkably improve pemetrexed antitumor activity in NSCLC patient-derived tumor mice, supporting the role of miR-7 in NSCLC metabolism and potential for BioRNA/miR-7 to improve NSCLC therapy.
Collapse
Affiliation(s)
- Gavin M Traber
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California
| | - Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California
| | - Su Guan
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California
| | - Neelu Batra
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California.
| |
Collapse
|
3
|
Cai R, Lin H, Cheng Q, Mao Q, Zhang C, Tan Y. Construction of a novel lipid drop-mitochondria-associated genetic profile for predicting the survival and prognosis of lung adenocarcinoma. Discov Oncol 2024; 15:668. [PMID: 39551861 PMCID: PMC11570572 DOI: 10.1007/s12672-024-01526-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is one of the most common malignant tumors. Although several treatments have been proposed, the long-term prognosis of this cancer is poor. Lipid droplets and mitochondria are important organelles that regulate energy metabolism in cells and are postulated to promote the occurrence and progression of tumors. However, few risk prediction models have been constructed based on lipid drop-mitochondria-related genes (LMRGs). METHODS In this study, we constructed a lipid drop-mitochondrial (LD-M) risk score model based on data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Biological functions and clinical benefits associated with the various risk scores were analyzed using R software, GraphPad Prism 9, and the online database system. RESULTS An LD-M risk score model comprising ABLIM3, AK4, CAV2, CPS1, CYP24A1, DLGAP5, FGR, and SH3BP5, was developed and its predictive power was validated. The risk score was closely associated with the cell cycle. Immunophenoscore (IPS) and Tumor immune dysfunction and exclusion (TIDE) results demonstrated that the low-risk group was more sensitive to immunotherapy. Drug sensitivity analysis indicated that BMS-754807, ZM447439, SB216763, and other drugs had lower IC50 values in the low-risk group. CONCLUSION Our results suggest that the LD-M risk score is an effective prognostic indicator for individualized treatment of LUAD.
Collapse
Affiliation(s)
- Ruijuan Cai
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongsheng Lin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | | | - Qiyuan Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuchu Zhang
- Institute of Chinese Medicine Information, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Tan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Sultana A, Alam MS, Khanam A, Lin Y, Ren S, Singla RK, Sharma R, Kuca K, Shen B. An integrated bioinformatics approach to early diagnosis, prognosis and therapeutics of non-small-cell lung cancer. J Biomol Struct Dyn 2024:1-15. [PMID: 39535278 DOI: 10.1080/07391102.2024.2425840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 05/31/2024] [Indexed: 11/16/2024]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most deadly tumors characterized by poor survival rates. Advances in therapeutics and precise identification of biomarkers can potentially reduce the mortality rate. Thus, this study aimed to identify a set of common and stable gene biomarkers through integrated bioinformatics approaches that might be effective for NSCLC early diagnosis, prognosis, and therapies. Four gene expression profiles (GSE19804, GSE19188, GSE10072, and GSE32863) downloaded from the Gene Expression Omnibus database to identify common differential expressed genes (DEGs). A total of 213 overlapping DEGs (oDEGs) between NSCLC and healthy samples were identified by using statistical LIMMA method. Then 6 common top-ranked key genes (KGs) (CENPF, CAV1, ASPM, CCNB2, PRC1, and KIAA0101) were selected by using four network-measurer methods in the protein- protein interaction network. The GO functional and KEGG pathway enrichment analysis were performed to reveal some significant functions and pathways associated with NSCLC progression. Transcriptional and post-transcriptional factors of KGs were identified through the regulatory interaction network. The prognostic power and expression level of KGs were validated by using the independent data through the Kaplan-Meier and Box plots, respectively. Finally, 4 KGs-guided repositioning candidate drugs (ZSTK474, GSK2126458, Masitinib, and Trametinib) were proposed. The stability of three top-ranked drug-target interactions (CAV1 vs. ZSTK474, CAV1 vs. GSK2126458, and ASPM vs. Trametinib) were investigated by computing their binding free energies for 140 ns MD-simulation based on MM-PBSA approach. Therefore, the findings of this computational study may be useful for early prognosis, diagnosis and therapies of NSCLC.
Collapse
Affiliation(s)
- Adiba Sultana
- School of Biology and Basic Medical Sciences, Soochow University Medical College, Suzhou, China
- Center for Systems Biology, Soochow University, Suzhou, China
- Medical Big Data Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Md Shahin Alam
- School of Biology and Basic Medical Sciences, Soochow University Medical College, Suzhou, China
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Alima Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Yuxin Lin
- Center for Systems Biology, Soochow University, Suzhou, China
| | - Shumin Ren
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Kamil Kuca
- Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Bairong Shen
- School of Biology and Basic Medical Sciences, Soochow University Medical College, Suzhou, China
- Center for Systems Biology, Soochow University, Suzhou, China
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Wang X, Ding R, Fu Z, Yang M, Li D, Zhou Y, Qin C, Zhang W, Si L, Zhang J, Chai Y. Overexpression of miR-506-3p reversed doxorubicin resistance in drug-resistant osteosarcoma cells. Front Pharmacol 2024; 15:1303732. [PMID: 38420199 PMCID: PMC10899521 DOI: 10.3389/fphar.2024.1303732] [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: 09/28/2023] [Accepted: 01/25/2024] [Indexed: 03/02/2024] Open
Abstract
Background and objective: Osteosarcoma is a common primary malignant tumor of bone, and doxorubicin is one of the most widely used therapeutic drugs. While the problem of doxorubicin resistance limits the long-term treatment benefits in osteosarcoma patients. The role of miRNAs and their target genes in osteosarcoma have become increasingly prominent. Currently, there is no report on miR-506-3p reversing doxorubicin resistance by targeting STAT3 in osteosarcoma. The purpose of this study was to investigate the molecular mechanism that overexpression of miR-506-3p reverses doxorubicin resistance in drug-resistant osteosarcoma cells. Methods: Doxorubicin-resistant osteosarcoma cells (U-2OS/Dox) were constructed by intermittent stepwise increasing stoichiometry. The target genes of miR-506-3p were predicted by bioinformatics approach and the targeting relationship between miR-506-3p and STAT3 was detected using dual luciferase reporter assay. U-2OS/Dox cells were treated with miR-506-3p overexpression and STAT3 silencing respectively. Then Western blot and RT-qPCR were used to detect the protein and mRNA expression levels of JAK2/STAT3 signaling pathway, drug-resistant and apoptotic associated molecules. The migration and invasion were assessed by cell scratch assay and transwell assay. The cell proliferative viability and apoptosis were investigated by CCK8 assay and flow cytometry assay. Results: U-2OS/Dox cells were successfully constructed with a 14.4-fold resistance. MiR-506-3p is directly bound to the 3'-UTR of STAT3 mRNA. Compared with U-2OS cells, the mRNA expression of miR-506-3p was reduced in U-2OS/Dox cells. Overexpression of miR-506-3p decreased the mRNA expression levels of JAK2, STAT3, MDR1/ABCB1, MRP1/ABCC1, Survivin and Bcl-2, and decreased the protein expression levels of p-JAK2, STAT3, MDR1/ABCB1, MRP1/ABCC1, Survivin and Bcl-2, and conversely increased Bax expression. It also inhibited the proliferation, migration and invasion of U-2OS/Dox cells and promoted cells apoptosis. The results of STAT3 silencing experiments in the above indicators were consistent with that of miR-506-3p overexpression. Conclusion: Overexpression of miR-506-3p could inhibit the JAK2/STAT3 pathway and the malignant biological behaviors, then further reverse doxorubicin resistance in drug-resistant osteosarcoma cells. The study reported a new molecular mechanism for reversing the resistance of osteosarcoma to doxorubicin chemotherapy and provided theoretical support for solving the clinical problems of doxorubicin resistance in osteosarcoma.
Collapse
Affiliation(s)
- Xinru Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rumeng Ding
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhe Fu
- Department of General Surgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Duolu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chongzhen Qin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenda Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liuzhe Si
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingmin Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuna Chai
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Sack T, Dhavarasa P, Szames D, O'Brien S, Angers S, Kelley SO. CRISPR Screening in Tandem with Targeted mtDNA Damage Reveals WRNIP1 Essentiality. ACS Chem Biol 2023; 18:2599-2609. [PMID: 38054633 DOI: 10.1021/acschembio.3c00620] [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: 12/07/2023]
Abstract
A major impediment to the characterization of mtDNA repair mechanisms in comparison to nuclear DNA repair mechanisms is the difficulty of specifically addressing mitochondrial damage. Using a mitochondria-penetrating peptide, we can deliver DNA-damaging agents directly to mitochondria, bypassing the nuclear compartment. Here, we describe the use of an mtDNA-damaging agent in tandem with CRISPR/Cas9 screening for the genome-wide discovery of factors essential for mtDNA damage response. Using mitochondria-targeted doxorubicin (mtDox), we generate mtDNA double-strand breaks (mtDSBs) specifically in this organelle. Combined with an untargeted doxorubicin (Dox) screen, we identify genes with significantly greater essentiality during mitochondrial versus nuclear DNA damage. We characterize the essentiality of our top hit, WRNIP1─observed here for the first time to respond to mtDNA damage. We further investigate the mitochondrial role of WRNIP1 in innate immune signaling and nuclear genome maintenance, outlining a model that experimentally supports mitochondrial turnover in response to mtDSBs.
Collapse
Affiliation(s)
- Tanja Sack
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Piriththiv Dhavarasa
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Daniel Szames
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Siobhan O'Brien
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Shana O Kelley
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Chemistry, Faculty of Arts and Science, University of Toronto, Toronto, Ontario M5S 3H6, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
- Chan Zuckerberg Biohub Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
7
|
Wan Y, Mu Q, Krzysztoń R, Cohen J, Coraci D, Helenek C, Tompkins C, Lin A, Farquhar K, Cross E, Wang J, Balázsi G. Adaptive DNA amplification of synthetic gene circuit opens a way to overcome cancer chemoresistance. Proc Natl Acad Sci U S A 2023; 120:e2303114120. [PMID: 38019857 DOI: 10.1073/pnas.2303114120] [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: 02/22/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Drug resistance continues to impede the success of cancer treatments, creating a need for experimental model systems that are broad, yet simple, to allow the identification of mechanisms and novel countermeasures applicable to many cancer types. To address these needs, we investigated a set of engineered mammalian cell lines with synthetic gene circuits integrated into their genome that evolved resistance to Puromycin. We identified DNA amplification as the mechanism underlying drug resistance in 4 out of 6 replicate populations. Triplex-forming oligonucleotide (TFO) treatment combined with Puromycin could efficiently suppress the growth of cell populations with DNA amplification. Similar observations in human cancer cell lines suggest that TFOs could be broadly applicable to mitigate drug resistance, one of the major difficulties in treating cancer.
Collapse
Affiliation(s)
- Yiming Wan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
| | - Quanhua Mu
- Department of Chemical and Biological Engineering, Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region 999077, China
| | - Rafał Krzysztoń
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
| | - Joseph Cohen
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
| | - Damiano Coraci
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
| | - Christopher Helenek
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
| | | | - Annie Lin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
| | - Kevin Farquhar
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
| | | | - Jiguang Wang
- Department of Chemical and Biological Engineering, Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region 999077, China
| | - Gábor Balázsi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794
| |
Collapse
|
8
|
Blair HC, Larrouture QC, Tourkova IL, Nelson DJ, Dobrowolski SF, Schlesinger PH. Epithelial-like transport of mineral distinguishes bone formation from other connective tissues. J Cell Biochem 2023; 124:1889-1899. [PMID: 37991446 PMCID: PMC10880123 DOI: 10.1002/jcb.30494] [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: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/24/2023] [Indexed: 11/23/2023]
Abstract
We review unique properties of bone formation including current understanding of mechanisms of bone mineral transport. We focus on formation only; mechanism of bone degradation is a separate topic not considered. Bone matrix is compared to other connective tissues composed mainly of the same proteins, but without the specialized mechanism for continuous transport and deposition of mineral. Indeed other connective tissues add mechanisms to prevent mineral formation. We start with the epithelial-like surfaces that mediate transport of phosphate to be incorporated into hydroxyapatite in bone, or in its ancestral tissue, the tooth. These include several phosphate producing or phosphate transport-related proteins with special expression in large quantities in bone, particularly in the bone-surface osteoblasts. In all connective tissues including bone, the proteins that constitute the protein matrix are mainly type I collagen and γ-carboxylate-containing small proteins in similar molar quantities to collagen. Specialized proteins that regulate connective tissue structure and formation are surprisingly similar in mineralized and non-mineralized tissues. While serum calcium and phosphate are adequate to precipitate mineral, specialized mechanisms normally prevent mineral formation except in bone, where continuous transport and deposition of mineral occurs.
Collapse
Affiliation(s)
- Harry C Blair
- Veteran’s Affairs Medical Center, Pittsburgh PA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | | | - Irina L. Tourkova
- Veteran’s Affairs Medical Center, Pittsburgh PA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Deborah J Nelson
- Dept of Neurobiology, Pharmacology & Physiology, University of Chicago, Chicago IL
| | | | | |
Collapse
|
9
|
Sack T, Dhavarasa P, Szames D, O'Brien S, Angers S, Kelley SO. CRISPR Screening in Tandem with Targeted mtDNA Damage Reveals WRNIP1 Essentiality. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560559. [PMID: 37873237 PMCID: PMC10592966 DOI: 10.1101/2023.10.03.560559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
A major impediment to the characterization of mtDNA repair mechanisms, in comparison to nuclear DNA repair mechanisms, is the difficulty of specifically addressing mitochondrial damage. Using a mitochondria-penetrating peptide, we can deliver DNA-damaging agents directly to mitochondria, bypassing the nuclear compartment. Here, we describe the use of a mtDNA-damaging agent in tandem with CRISPR/Cas9 screening for the genome-wide discovery of factors essential for mtDNA damage response. Using mitochondria-targeted doxorubicin (mtDox) we generate mtDNA double-strand breaks (mtDSBs) specifically in this organelle. Combined with an untargeted Dox screen, we identify genes with significantly greater essentiality during mitochondrial versus nuclear DNA damage. We characterize the essentially of our top hit - WRNIP1 - observed here for the first time to respond to mtDNA damage. We further investigate the mitochondrial role of WRNIP1 in innate immune signaling and nuclear genome maintenance, outlining a model that experimentally supports mitochondrial turnover in response to mtDSBs.
Collapse
|
10
|
Boichuk S, Bikinieva F, Valeeva E, Dunaev P, Vasileva M, Kopnin P, Mikheeva E, Ivoilova T, Mustafin I, Galembikova A. Establishment and Characterization of Multi-Drug Resistant p53-Negative Osteosarcoma SaOS-2 Subline. Diagnostics (Basel) 2023; 13:2646. [PMID: 37627905 PMCID: PMC10453552 DOI: 10.3390/diagnostics13162646] [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: 07/14/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
AIM To establish a p53-negative osteosarcoma (OS) SaOS-2 cellular subline exhibiting resistance to specific chemotherapeutic agents, including topoisomerase II inhibitors, taxanes, and vinca alkaloids. METHODS The OS subline exhibiting resistance to the chemotherapeutic agents indicated above was generated by the stepwise treatment of the parental SaOS-2 cell line with increasing concentrations of doxorubicin (Dox) for 5 months. Half-inhibitory concentrations (IC50) for Dox, vinblastine (Vin), and paclitaxel (PTX) were calculated by a colorimetric MTS-based assay. Crystal violet staining was used to assess cellular viability, whereas the proliferation capacities of cancer cells were monitored in real-time by the i-Celligence system. Expression of apoptotic markers (e.g., cleaved PARP and caspase-3), DNA repair proteins (e.g., ATM, DNA-PK, Nbs1, Rad51, MSH2, etc.), and certain ABC transporters (P-glycoprotein, MRP1, ABCG2, etc.) was assessed by western blotting and real-time PCR. Flow cytometry was used to examine the fluorescence intensity of Dox and ABC-transporter substrates (e.g., Calcein AM and CMFDA) and to assess their excretion to define the activity of specific ABC-transporters. To confirm OS resistance to Dox in vivo, xenograft experiments were performed. RESULTS An OS subline generated by a stepwise treatment of the parental SaOS-2 cell line with increasing concentrations of Dox resulted in an increase in the IC50 for Dox, Vin, and PTX (~6-, 4-, and 30-fold, respectively). The acquisition of chemoresistance in vitro was also evidenced by the lack of apoptotic markers (e.g., cleaved PARP and caspase-3) in resistant OS cells treated with the chemotherapeutic agents indicated above. The development of the multidrug resistance (MDR) phenotype in this OS subline was due to the overexpression of ABCB1 (i.e., P-glycoprotein) and ABCC1 (i.e., multidrug resistance protein-1, MRP-1), which was evidenced on both mRNA and protein levels. Due to increased expression of MDR-related proteins, resistant OS exhibited an excessive efflux of Dox. Moreover, decreased accumulation of calcein AM, a well-known fluorescent substrate for both ABCB1 and ABCC1, was observed for resistant OS cells compared to their parental SaOS-2 cell line. Importantly, tariquidar and cyclosporin, well-known ABC inhibitors, retained the intensity of Dox-induced fluorescence in resistant SAOS-2 cells. Furthermore, in addition to the increased efflux of the chemotherapeutic agents from Dox-resistant OS cells, we found higher expression of several DNA repair proteins (e.g., Rad51 recombinase, Mre11, and Nbs1, activated forms of ATM, DNA-PK, Chk1, and Chk2, etc.), contributing to the chemoresistance due to the excessive DNA repair. Lastly, the in vivo study indicated that Dox has no impact on the SaOS-2 Dox-R xenograft tumor growth in a nude mouse model. CONCLUSIONS An acquired resistance of OS to the chemotherapeutic agents might be due to the several mechanisms undergoing simultaneously on the single-cell level. This reveals the complexity of the mechanisms involved in the secondary resistance of OS to chemotherapies.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (F.B.); (P.D.); (E.M.); (T.I.); (A.G.)
- ”Biomarker” Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Department of Radiotherapy and Radiology, Russian Medical Academy of Continuous Professional Education, Moscow 125993, Russia
| | - Firyuza Bikinieva
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (F.B.); (P.D.); (E.M.); (T.I.); (A.G.)
| | - Elena Valeeva
- Central Research Laboratory, Kazan State Medical University, Kazan 420012, Russia;
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (F.B.); (P.D.); (E.M.); (T.I.); (A.G.)
| | - Maria Vasileva
- Cytogenetics Laboratory, Carcinogenesis Institute, N.N. Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; (M.V.); (P.K.)
| | - Pavel Kopnin
- Cytogenetics Laboratory, Carcinogenesis Institute, N.N. Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; (M.V.); (P.K.)
| | - Ekaterina Mikheeva
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (F.B.); (P.D.); (E.M.); (T.I.); (A.G.)
| | - Tatyana Ivoilova
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (F.B.); (P.D.); (E.M.); (T.I.); (A.G.)
| | - Ilshat Mustafin
- Department of Biochemistry, Kazan State Medical University, Kazan 420012, Russia;
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (F.B.); (P.D.); (E.M.); (T.I.); (A.G.)
| |
Collapse
|
11
|
de Almeida Roque A, da Luz JZ, Santurio MTK, Neto FF, de Oliveira Ribeiro CA. Complex mixtures of pesticides and metabolites modulate the malignant phenotype of murine melanoma B16-F1 cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:47366-47380. [PMID: 36738412 DOI: 10.1007/s11356-023-25603-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Pesticides use increased worldwide with a record in Brazil. Although several works addressed the effects of pesticides on living organisms, only a few considered their mixture, and even fewer tried to unravel their role in tumoral progression. Due to the relevance of cancer, in the present study, the effects of the mixture of pesticides widely used in Brazil (Glyphosate, 2,4-dichlorophenoxyacetic acid, Mancozeb, Atrazine, Acephate, and Paraquat) and their main metabolites (Aminomethylphosphonic Acid, 2,4-diclorophenol, Ethylenethiourea, Desethylatrazine, Methamidophos, and Paraquat) were investigated on the malignancy phenotype of murine melanoma B16-F1 cells after acute (24 h) and chronic (15 days) exposures. The tested concentrations were based on the Acceptable Daily Intake (ADI) value established by Brazilian legislation. The set of results showed that these chemicals modulate important parameters of tumor progression, affecting the expression of genes related to tumor aggressiveness (Mmp14 and Cd44) and multidrug resistance (Abcb1, Abcc1, and Abcc4), as well as tissue inhibitors of metalloproteinases (Timp1, Timp2, and Timp3). These findings revealed an absence of cytotoxicity but showed modulation of migration, invasion, and colonization capacity of B16-F1 cells. Together, the results point to some negative ways that exposure to pesticides can affect the progression of melanoma and raise a concern related to the increasing trend in pesticide use in Brazil, as the country is one of the major world food suppliers.
Collapse
Affiliation(s)
- Aliciane de Almeida Roque
- Laboratory of Cell Toxicology, Department of Cellular and Molecular Biology, Federal University of Paraná, PO Box: 19031, Curitiba, PR, CEP: 81531-980, Brazil
| | - Jessica Zablocki da Luz
- Laboratory of Cell Toxicology, Department of Cellular and Molecular Biology, Federal University of Paraná, PO Box: 19031, Curitiba, PR, CEP: 81531-980, Brazil
| | - Michelle Thays Khun Santurio
- Laboratory of Cell Toxicology, Department of Cellular and Molecular Biology, Federal University of Paraná, PO Box: 19031, Curitiba, PR, CEP: 81531-980, Brazil
| | - Francisco Filipak Neto
- Laboratory of Cell Toxicology, Department of Cellular and Molecular Biology, Federal University of Paraná, PO Box: 19031, Curitiba, PR, CEP: 81531-980, Brazil
| | - Ciro Alberto de Oliveira Ribeiro
- Laboratory of Cell Toxicology, Department of Cellular and Molecular Biology, Federal University of Paraná, PO Box: 19031, Curitiba, PR, CEP: 81531-980, Brazil.
| |
Collapse
|
12
|
Semi-Synthetic Dihydrotestosterone Derivatives Modulate Inherent Multidrug Resistance and Sensitize Colon Cancer Cells to Chemotherapy. Pharmaceutics 2023; 15:pharmaceutics15020584. [PMID: 36839907 PMCID: PMC9966060 DOI: 10.3390/pharmaceutics15020584] [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: 12/07/2022] [Revised: 01/21/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Multidrug resistance (MDR) is a serious hurdle to successful cancer therapy. Here, we examined the efficiency of novel semi-synthetic dihydrotestosterone derivatives, more specifically androstano-arylpyrimidines in inhibiting the efflux activity of ATP-binding cassette (ABC) transporters and sensitizing inherently MDR colon cancer cells to various chemotherapy drugs. Using the Rhodamine123 accumulation assay, we evaluated the efflux activity of cancer cells following treatments with androstano-arylpyrimidines. We found that acetylated compounds were capable of attenuating the membrane efflux of inherently MDR cells; however, deacetylated counterparts were ineffective. To delineate the possible molecular mechanisms underlying these unique activities of androstano-arylpyrimidines, the degree of apoptosis induction was assessed by AnnexinV-based assays, both upon the individual as well as by steroid and chemotherapy agent combination treatments. Five dihydrotestosterone derivatives applied in combination with Doxorubicin or Epirubicin triggered massive apoptosis in MDR cells, and these combinations were more efficient than chemotherapy drugs together with Verapamil. Furthermore, our results revealed that androstano-arylpyrimidines induced significant endoplasmic reticulum stress (ER stress) but did not notably modulate ABC transporter expression. Therefore, ER stress triggered by acetylated androstano-arylpyrimidines is probably involved in the mechanism of efflux pump inhibition and drug sensitization which can be targeted in future drug developments to defeat inherently multidrug-resistant cancer.
Collapse
|
13
|
Zhao Y, Tang C, Huang J, Zhang H, Shi J, Xu S, Ma L, Peng C, Liu Q, Xiong Y. Screening Multidrug Resistance Reversal Agents in Traditional Chinese Medicines by Efflux Kinetics of D-Luciferin in MCF-7/DOX Fluc Cells. ACS OMEGA 2023; 8:4853-4861. [PMID: 36777569 PMCID: PMC9909823 DOI: 10.1021/acsomega.2c07096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/05/2023] [Indexed: 06/18/2023]
Abstract
In this study, we established a simple and rapid in vitro method for screening multidrug resistance (MDR) reversal agents in traditional Chinese medicines (TCMs), which could better correspond to the MDR reversing effect in vivo. Here, D-luciferin, a substrate for the enzyme firefly luciferase and also a substrate for ATP-binding cassette transporters (ABC transporters), was used as the probe to detect its efflux kinetics caused by ABC transporters. First, we established a stable doxorubicin (DOX)-resistant cell line (MCF-7/DOXFluc) that overexpressed luciferase. Then, some kinds of TCMs were chosen for the MDR reversal agents to measure its effect on inhibiting the D-luciferin outflow from MCF-7/DOXFluc, and the ideal reversal agent with the least D-luciferin efflux from MCF-7/DOXFluc was selected to further investigate its effect combined with DOX on MCF-7/DOXFluc tumor-bearing mice. The results indicated that quercetin (Qu) could remarkably increase the retention of D-luciferin in MCF-7/DOXFluc in vitro and in vivo. Also, the combination of Qu and DOX could exceedingly inhibit the tumor growth, which proved the feasibility of this in vitro screening method. The study proposed a feasible method for mass screening of MDR agents from TCMs in vitro.
Collapse
Affiliation(s)
- Yue Zhao
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
- Academy
of Chinese Medical Science, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
| | - Chaoyuan Tang
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
- Changxing
People’s Hospital of Zhejiang, Huzhou, Zhejiang 313100, China
| | - Jingyi Huang
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
| | - Hongyan Zhang
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
| | - Jingbin Shi
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
| | - Shujun Xu
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
| | - Lisha Ma
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
| | - Chun Peng
- School
of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Qi Liu
- Department
of Dermatology, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21231, United States
| | - Yang Xiong
- College
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
- Academy
of Chinese Medical Science, Zhejiang Chinese
Medical University, Hangzhou, Zhejiang 311258, China
| |
Collapse
|
14
|
Shivhare S, Das A. Cell density modulates chemoresistance in breast cancer cells through differential expression of ABC transporters. Mol Biol Rep 2023; 50:215-225. [PMID: 36319789 DOI: 10.1007/s11033-022-08028-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/12/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Breast cancer patients undergoing chemotherapy encounter a significant challenge of chemoresistance because of drug efflux by ATP-binding cassette (ABC) transporters. Breast cancer cell density alters considerably throughout the early stages of primary and secondary tumor development. Although cell density in culture influences kinetics, the effects of varying cell densities on the chemoresistance of breast cancer cells remains largely unexplored. METHODS AND RESULTS We observed chemotherapeutics-induced differential gene and protein expression of ABC transporters in luminal and basal breast cancer cells cultured at low and high seeding densities. Low-density cultures depicted a significant increase in the mRNA expression of ABC transporters-ABCG2, ABCG1, ABCC4, ABCA2, ABCA3, ABCC2, ABCC3, ABCC6, ABCC7, and ABCC9 as compared with high-density cultures. Next, cells at both low and high seeding densities when pre-treated with cyclosporine A (CsA), a pan-inhibitor of ABC transporters, resulted in increased sensitization to chemotherapeutics-doxorubicin and tamoxifen at markedly low IC50 concentrations suggesting the role of ABC transporters. Finally, markedly high doxorubicin-drug accumulation, significantly increased expression of N-cadherin, and a significant decrease in chemotherapeutics-induced in vitro tumorigenesis was observed in low-density seeded breast cancer cells when pre-treated with CsA suggesting ABC transporters inhibition-mediated increased efficacy of chemotherapeutics. CONCLUSION These findings suggest that breast cancer cells grown at low seeding density imparts chemoresistance towards doxorubicin or tamoxifen by a differential increase in the expression of ABC transporters. Thus, a combinatorial treatment strategy including ABC transporter inhibitors and chemotherapeutics can be a way forward for overcoming the breast cancer chemoresistance.
Collapse
Affiliation(s)
- Surbhi Shivhare
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, TS, 500 007, India.,Academy of Scientific and Innovative Research, Ghaziabad, UP, 201 002, India
| | - Amitava Das
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, TS, 500 007, India. .,Academy of Scientific and Innovative Research, Ghaziabad, UP, 201 002, India.
| |
Collapse
|
15
|
Rösch L, Herter S, Najafi S, Ridinger J, Peterziel H, Cinatl J, Jones DTW, Michaelis M, Witt O, Oehme I. ERBB and P-glycoprotein inhibitors break resistance in relapsed neuroblastoma models through P-glycoprotein. Mol Oncol 2022; 17:37-58. [PMID: 36181342 PMCID: PMC9812835 DOI: 10.1002/1878-0261.13318] [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: 08/26/2022] [Accepted: 09/29/2022] [Indexed: 02/03/2023] Open
Abstract
Chemotherapy resistance is a persistent clinical problem in relapsed high-risk neuroblastomas. We tested a panel of 15 drugs for sensitization of neuroblastoma cells to the conventional chemotherapeutic vincristine, identifying tariquidar, an inhibitor of the transmembrane pump P-glycoprotein (P-gp/ABCB1), and the ERBB family inhibitor afatinib as the top resistance breakers. Both compounds were efficient in sensitizing neuroblastoma cells to vincristine in trypan blue exclusion assays and in inducing apoptotic cell death. The evaluation of ERBB signaling revealed no functional inhibition, that is, dephosphorylation of the downstream pathways upon afatinib treatment but direct off-target interference with P-gp function. Depletion of ABCB1, but not ERRB4, sensitized cells to vincristine treatment. P-gp inhibition substantially broke vincristine resistance in vitro and in vivo (zebrafish embryo xenograft). The analysis of gene expression datasets of more than 50 different neuroblastoma cell lines (primary and relapsed) and more than 160 neuroblastoma patient samples from the pediatric precision medicine platform INFORM (Individualized Therapy For Relapsed Malignancies in Childhood) confirmed a pivotal role of P-gp specifically in neuroblastoma resistance at relapse, while the ERBB family appears to play a minor part.
Collapse
Affiliation(s)
- Lisa Rösch
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Clinical Cooperation Unit Pediatric OncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany,Faculty of BiosciencesUniversity of HeidelbergGermany
| | - Sonja Herter
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Clinical Cooperation Unit Pediatric OncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany,Faculty of BiosciencesUniversity of HeidelbergGermany
| | - Sara Najafi
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Clinical Cooperation Unit Pediatric OncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany,Department of Pediatric Oncology, Hematology and ImmunologyUniversity Hospital HeidelbergGermany
| | - Johannes Ridinger
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Clinical Cooperation Unit Pediatric OncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| | - Heike Peterziel
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Clinical Cooperation Unit Pediatric OncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| | - Jindrich Cinatl
- Institute for Medical VirologyGoethe University HospitalFrankfurt am MainGermany
| | - David T. W. Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Division of Pediatric Glioma ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Clinical Cooperation Unit Pediatric OncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany,Department of Pediatric Oncology, Hematology and ImmunologyUniversity Hospital HeidelbergGermany
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ)Germany,Clinical Cooperation Unit Pediatric OncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| |
Collapse
|
16
|
Zhang L, Ye B, Chen Z, Chen ZS. Progress in the studies on the molecular mechanisms associated with multidrug resistance in cancers. Acta Pharm Sin B 2022; 13:982-997. [PMID: 36970215 PMCID: PMC10031261 DOI: 10.1016/j.apsb.2022.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/28/2022] [Accepted: 08/18/2022] [Indexed: 11/01/2022] Open
Abstract
Chemotherapy is one of the important methods to treat cancer, and the emergence of multidrug resistance (MDR) is one major cause for the failure of cancer chemotherapy. Almost all anti-tumor drugs develop drug resistance over a period of time of application in cancer patients, reducing their effects on killing cancer cells. Chemoresistance can lead to a rapid recurrence of cancers and ultimately patient death. MDR may be induced by multiple mechanisms, which are associated with a complex process of multiple genes, factors, pathways, and multiple steps, and today the MDR-associated mechanisms are largely unknown. In this paper, from the aspects of protein-protein interactions, alternative splicing (AS) in pre-mRNA, non-coding RNA (ncRNA) mediation, genome mutations, variance in cell functions, and influence from the tumor microenvironment, we summarize the molecular mechanisms associated with MDR in cancers. In the end, prospects for the exploration of antitumor drugs that can reverse MDR are briefly discussed from the angle of drug systems with improved targeting properties, biocompatibility, availability, and other advantages.
Collapse
|
17
|
Ceramide Metabolism Regulated by Sphingomyelin Synthase 2 Is Associated with Acquisition of Chemoresistance via Exosomes in Human Leukemia Cells. Int J Mol Sci 2022; 23:ijms231810648. [PMID: 36142562 PMCID: PMC9505618 DOI: 10.3390/ijms231810648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/07/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
Ceramide levels controlled by the sphingomyelin (SM) cycle have essential roles in cancer cell fate through the regulation of cell proliferation, death, metastasis, and drug resistance. Recent studies suggest that exosomes confer cancer malignancy. However, the relationship between ceramide metabolism and exosome-mediated cancer malignancy is unclear. In this study, we elucidated the role of ceramide metabolism via the SM cycle in exosomes and drug resistance in human leukemia HL-60 and adriamycin-resistant HL-60/ADR cells. HL-60/ADR cells showed significantly increased exosome production and release compared with parental chemosensitive HL-60 cells. In HL-60/ADR cells, increased SM synthase (SMS) activity reduced ceramide levels, although released exosomes exhibited a high ceramide ratio in both HL-60- and HL-60/ADR-derived exosomes. Overexpression of SMS2 but not SMS1 suppressed intracellular ceramide levels and accelerated exosome production and release in HL-60 cells. Notably, HL-60/ADR exosomes conferred cell proliferation and doxorubicin resistance properties to HL-60 cells. Finally, microRNA analysis in HL-60 and HL-60/ADR cells and exosomes showed that miR-484 elevation in HL-60/ADR cells and exosomes was associated with exosome-mediated cell proliferation. This suggests that intracellular ceramide metabolism by SMS2 regulates exosome production and release, leading to acquisition of drug resistance and enhanced cell proliferation in leukemia cells.
Collapse
|
18
|
Looking at NSAIDs from a historical perspective and their current status in drug repurposing for cancer treatment and prevention. J Cancer Res Clin Oncol 2022; 149:2095-2113. [PMID: 35876951 PMCID: PMC9310000 DOI: 10.1007/s00432-022-04187-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most frequently prescribed drug classes with wide therapeutic applications over the centuries. Starting from the use of salicylate-containing willow leaves to the recent rise and fall of highly selective cyclooxygenase-2 (COX-2) inhibitors and the latest dual-acting anti-inflammatory molecules, they have displayed a rapid and ongoing evolution. Despite the enormous advances in the last twenty years, investigators are still in search of the design and development of more potent and safer therapy against inflammatory conditions. This challenge has been increasingly attractive as the emergence of inflammation as a common seed and unifying mechanism for most chronic diseases. Indeed, this fact put the NSAIDs in the spotlight for repurposing against inflammation-related disorders. This review attempts to present a historical perspective on the evolution of NSAIDs, regarding their COX-dependent/independent mode of actions, structural and mechanism-based classifications, and adverse effects. Additionally, a systematic review of previous studies was carried out to show the current situation in drug repurposing, particularly in cancers associated with the GI tract such as gastric and colorectal carcinoma. In the case of non-GI-related cancers, preclinical studies elucidating the effects and modes of action were collected and summarized.
Collapse
|
19
|
Shukla SK, Sarode A, Wang X, Mitragotri S, Gupta V. Particle shape engineering for improving safety and efficacy of doxorubicin - A case study of rod-shaped carriers in resistant small cell lung cancer. BIOMATERIALS ADVANCES 2022; 137:212850. [PMID: 35929278 DOI: 10.1016/j.bioadv.2022.212850] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/14/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022]
Abstract
Therapeutic drug delivery is known to be influenced by interplay between various design parameters of delivery carriers which influence the drug uptake efficiency and subsequently the effectiveness of treatment. Amongst, the several design parameters such as size, shape and surface charge, particle shape is gaining attention as a crucial design parameter for development of robust and efficient delivery carriers. In this exploration, we investigated the influence of particle shape on injectability and therapeutic effectiveness of the delivery carriers using doxorubicin (DOX) conjugated polymeric microparticles. Results of injectability experiments demonstrated the influence of particle shape with anisotropic rod-shaped particles displaying increased injectability as against spherical particles. Impact of particle shape on therapeutic effectiveness was assessed against small cell lung cancer (SCLC) which was selected as a model disease. Results of cellular uptake studies revealed preferential uptake of rod-shaped particles than spherical particles in cancer cells. These results were further validated by in-vitro tumor simulation studies wherein rod-shaped particles displayed enhanced anti-tumorigenic activity along with distortion of tumor integrity against spheres. Furthermore, the impact of particle size was also assessed on cardiotoxicity, an adverse effect of DOX which limits its therapeutic use. Results illustrated that the high aspect ratio particles displayed diminished cardiotoxicity activity. These results provide valuable insights about influence of particle shape for designing efficient therapeutics.
Collapse
Affiliation(s)
- Snehal K Shukla
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Apoorva Sarode
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Xuechun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Samir Mitragotri
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
20
|
Poku VO, Iram SH. A critical review on modulators of Multidrug Resistance Protein 1 in cancer cells. PeerJ 2022; 10:e12594. [PMID: 35036084 PMCID: PMC8742536 DOI: 10.7717/peerj.12594] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/14/2021] [Indexed: 01/11/2023] Open
Abstract
Multidrug resistance protein 1 (MRP1/ABCC1) is an ATP-dependent efflux transporter, and responsible for the transport of a broad spectrum of xenobiotics, toxins, and physiological substrates across the plasma membrane. As an efflux pump, it plays a significant role in the absorption and disposition of drugs including anticancer drugs, antivirals, antimalarials, and antibiotics and their metabolites across physiological barriers in cells. MRP1 is also known to aid in the regulation of several physiological processes such as redox homeostasis, steroid metabolism, and tissue defense. However, its overexpression has been reported to be a key clinical marker associated with multidrug resistance (MDR) of several types of cancers including lung cancer, childhood neuroblastoma, breast and prostate carcinomas, often resulting in a higher risk of treatment failure and shortened survival rates in cancer patients. Aside MDR, overexpression of MRP1 is also implicated in the development of neurodegenerative and cardiovascular diseases. Due to the cellular importance of MRP1, the identification and biochemical/molecular characterization of modulators of MRP1 activity and expression levels are of key interest to cancer research and beyond. This review primarily aims at highlighting the physiological and pharmacological importance of MRP1, known MRP1 modulators, current challenges encountered, and the potential benefits of conducting further research on the MRP1 transporter.
Collapse
Affiliation(s)
- Vivian Osei Poku
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States of America
| | - Surtaj Hussain Iram
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States of America,American University of Iraq, Sulaimaniya, Sulaimani, KRG, Iraq
| |
Collapse
|
21
|
Ostlund T, Alotaibi F, Kyeremateng J, Halaweish H, Kasten A, Iram S, Halaweish F. Triazole-estradiol analogs: A potential cancer therapeutic targeting ovarian and colorectal cancer. Steroids 2022; 177:108950. [PMID: 34933058 DOI: 10.1016/j.steroids.2021.108950] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 11/08/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023]
Abstract
1,2,3-triazoles have continuously shown effectiveness as biologically active systems towards various cancers, and when used in combination with steroid skeletons as a carrier, which can act as a drug delivery system, allows for a creation of a novel set of analogs that may be useful as a pharmacophore leading to a potential treatment option for cancer. A common molecular target for cancer inhibition is that of the Epidermal Growth Factor Receptor/Mitogen Activated Protein Kinase pathways, as inhibition of these proteins is associated with a decrease in cell viability. Estradiol-Triazole analogs were thus designed using a molecular modeling approach. Thirteen of the high scoring analogs were then synthesized and tested in-vitro on an ovarian cancer cell line (A2780) and colorectal cancer cell line (HT-29). The most active compound, Fz25, shows low micromolar activity in both the ovarian (15.29 ± 2.19 µM) and colorectal lines (15.98 ± 0.39 µM). Mechanism of action studies proved that Fz25 moderately arrests cells in the G1 phase of the cell cycle, specifically inhibiting STAT3 in both cell lines. Additionally, Fz57 shows activity in the colorectal line (24.19 ± 1.37 µM). Inhibition studies in both cell lines show inhibition against various proteins in the EGFR pathway, namely EGFR, STAT3, ERK, and mTOR. To further study their effects as therapeutics, Fz25 and Fz57 were studied against drug efflux proteins, which are associated with drug resistance, and were found to inhibit the ABC transporter P-glycoprotein. We can conclude that these estradiol-triazole analogs provide a key for future studies targeting protein inhibition and drug resistance in cancer.
Collapse
Affiliation(s)
- Trevor Ostlund
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007, United States
| | - Faez Alotaibi
- Department of Chemistry & Biochemistry, North Dakota State University, Fargo, ND 58105, United States
| | - Jennifer Kyeremateng
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007, United States
| | - Hossam Halaweish
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota, 420 Delaware St SE. MMC 195, Minneapolis, MN 55455, United States
| | - Abigail Kasten
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007, United States
| | - Surtaj Iram
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007, United States
| | - Fathi Halaweish
- Department of Chemistry & Biochemistry, South Dakota State University, Brookings, SD 57007, United States.
| |
Collapse
|
22
|
Hanssen KM, Haber M, Fletcher JI. Targeting multidrug resistance-associated protein 1 (MRP1)-expressing cancers: Beyond pharmacological inhibition. Drug Resist Updat 2021; 59:100795. [PMID: 34983733 DOI: 10.1016/j.drup.2021.100795] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 12/30/2022]
Abstract
Resistance to chemotherapy remains one of the most significant obstacles to successful cancer treatment. While inhibiting drug efflux mediated by ATP-binding cassette (ABC) transporters is a seemingly attractive and logical approach to combat multidrug resistance (MDR), small molecule inhibition of ABC transporters has so far failed to confer clinical benefit, despite considerable efforts by medicinal chemists, biologists, and clinicians. The long-sought treatment to eradicate cancers displaying ABC transporter overexpression may therefore lie within alternative targeting strategies. When aberrantly expressed, the ABC transporter multidrug resistance-associated protein 1 (MRP1, ABCC1) confers MDR, but can also shift cellular redox balance, leaving the cell vulnerable to select agents. Here, we explore the physiological roles of MRP1, the rational for targeting this transporter in cancer, the development of small molecule MRP1 inhibitors, and the most recent developments in alternative therapeutic approaches for targeting cancers with MRP1 overexpression. We discuss approaches that extend beyond simple MRP1 inhibition by exploiting the collateral sensitivity to glutathione depletion and ferroptosis, the rationale for targeting the shared transcriptional regulators of both MRP1 and glutathione biosynthesis, advances in gene silencing, and new molecules that modulate transporter activity to the detriment of the cancer cell. These strategies illustrate promising new approaches to address multidrug resistant disease that extend beyond the simple reversal of MDR and offer exciting routes for further research.
Collapse
Affiliation(s)
- Kimberley M Hanssen
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
23
|
Mynott RL, Wallington-Beddoe CT. Drug and Solute Transporters in Mediating Resistance to Novel Therapeutics in Multiple Myeloma. ACS Pharmacol Transl Sci 2021; 4:1050-1065. [PMID: 34151200 DOI: 10.1021/acsptsci.1c00074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 02/06/2023]
Abstract
Multiple myeloma remains an incurable malignancy of plasma cells. Novel therapies, notably proteasome inhibitors and immunomodulatory drugs, have improved the survival of multiple myeloma patients; however, patients either present with, or develop resistance to, these therapies. Resistance to traditional chemotherapeutic agents can be caused by cellular drug efflux via adenosine triphosphate (ATP)-binding cassette (ABC) transporters, but it is still not clear whether these transporters mediate resistance to proteasome inhibitors and immunomodulatory drugs in multiple myeloma. Solute carrier (SLC) transporters also play a role in cancer drug resistance due to changes in cell homeostasis caused by their abnormal expression and changes in the solutes they transport. In this review, we evaluate resistance to novel therapies used to treat multiple myeloma, as mediated by drug and solute transporters.
Collapse
Affiliation(s)
- Rachel L Mynott
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Craig T Wallington-Beddoe
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia.,Flinders Medical Centre, Bedford Park, South Australia 5042, Australia.,Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, South Australia 5000, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| |
Collapse
|
24
|
Shawky AM, Abdalla AN, Ibrahim NA, Abourehab MAS, Gouda AM. Discovery of new pyrimidopyrrolizine/indolizine-based derivatives as P-glycoprotein inhibitors: Design, synthesis, cytotoxicity, and MDR reversal activities. Eur J Med Chem 2021; 218:113403. [PMID: 33823396 DOI: 10.1016/j.ejmech.2021.113403] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/11/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022]
Abstract
Targeting P-glycoprotein (P-gp, ABCB1 transporter), which plays an essential role in multi-drug resistance (MDR) in cancers, with new cytotoxic agents is a promising strategy in cancer chemotherapy. In the current study, we report the synthesis of thirteen novel pyrimidopyrrolizine, pyrimidoindolizine, and diazepinopyrrolizine derivatives. The new compounds exhibited cytotoxic activities against MCF7, A2780 and HT29 cancer cell lines (IC50 = 0.02-19.58 μM) and MRC5 (IC50 = 0.17-20.57 μM). The six most active compounds (23b, 24a,b and, 31c-e) were evaluated for their MDR reversal activities in MCF7/ADR cells. Mechanistic study using real-time PCR revealed the ability of compound 31c to inhibit P-gp. In addition, compound 31c increased the accumulation of Rho123 inside MCF7/ADR cells in a dose-dependent manner compared to verapamil. Compound 31c arrested the cell cycle of MCF7 cells at the G1 phase. Compound 31c also caused a significant dose-dependent increase of early and late apoptotic events. Molecular docking analysis revealed a high binding affinity for compound 31c toward P-gp. Like zosuquidar, compound 31c displayed one hydrogen bond and several hydrophobic interactions with amino acids in P-gp. These results indicated that compound 31c represents a potential anticancer candidate with MDR reversal activity.
Collapse
Affiliation(s)
- Ahmed M Shawky
- Science and Technology Unit (STU), Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Central Laboratory for Micro-analysis, Minia University, Minia, 61519, Egypt
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Departmentof Pharmacology and Toxicology, Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum, 2404, Sudan
| | - Nashwa A Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Ahmed M Gouda
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
25
|
Juan-Carlos PDM, Perla-Lidia PP, Stephanie-Talia MM, Mónica-Griselda AM, Luz-María TE. ABC transporter superfamily. An updated overview, relevance in cancer multidrug resistance and perspectives with personalized medicine. Mol Biol Rep 2021; 48:1883-1901. [PMID: 33616835 DOI: 10.1007/s11033-021-06155-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/12/2021] [Indexed: 12/25/2022]
Abstract
The ATP binding-cassette superfamily corresponds the mostly transmembrane transporters family found in humans. These proteins actively transport endogenous and exogenous substrates through biological membranes in body tissues, so they have an important role in the regulation of many physiological functions necessary for human homeostasis, as well as in response regulation to several pharmacological substrates. The development of multidrug resistance has become one of the main troubles in conventional chemotherapy in different illnesses including cancer, being the increased efflux of antineoplastic drugs the main reason for this multidrug resistance, with a key role of the ABC superfamily. Likely, the interindividual variability in the pharmacological response among patients is well known, and may be due to intrinsically factors of the disease, genetic and environmental ones. Thus, the understanding of this variability, especially the genetic variability associated with the efficacy and toxicity of drugs, can provide a safer and more effective pharmacological treatment, so ABC genes are considered as important regulators due to their relationship with the reduction in pharmacological response. In this review, updated information about transporters belonging to this superfamily was collected, the possible role of these transporters in cancer, the role of genetic variability in their genes, as well as some therapeutic tools that have been tried to raise against main transporters associated with chemoresistance in cancer.
Collapse
Affiliation(s)
- Pérez-De Marcos Juan-Carlos
- Laboratory of Pharmacology, National Institute of Pediatrics, Mexico City, México.,Postgraduate Degree in Pharmacology, National Polytechnic Institute, Mexico City, México
| | | | | | | | | |
Collapse
|
26
|
Alim K, Bruyère A, Lescoat A, Jouan E, Lecureur V, Le Vée M, Fardel O. Interactions of janus kinase inhibitors with drug transporters and consequences for pharmacokinetics and toxicity. Expert Opin Drug Metab Toxicol 2021; 17:259-271. [PMID: 33292029 DOI: 10.1080/17425255.2021.1862084] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Janus kinase inhibitors (JAKinibs) constitute an emerging and promising pharmacological class of anti-inflammatory or anti-cancer drugs, used notably for the treatment of rheumatoid arthritis and some myeloproliferative neoplasms.Areas covered: This review provides an overview of the interactions between marketed JAKinibs and major uptake and efflux drug transporters. Consequences regarding pharmacokinetics, drug-drug interactions and toxicity are summarized.Expert opinion: JAKinibs interact in vitro with transporters in various ways, as inhibitors or as substrates of transporters or as regulators of transporter expression. This may theoretically result in drug-drug interactions (DDIs), with JAKinibs acting as perpetrators or as victims, or in toxicity, via impairment of thiamine transport. Clinical significance in terms of DDIs for JAKinib-transporter interactions remains however poorly documented. In this context, the in vivo unbound concentration of JAKinibs is likely a key parameter to consider for evaluating the clinical relevance of JAKinibs-mediated transporter inhibition. Additionally, the interplay with drug metabolism as well as possible interactions with transporters of emerging importance and time-dependent inhibition have to be taken into account. The role drug transporters may play in controlling cellular JAKinib concentrations and efficacy in target cells is also an issue of interest.
Collapse
Affiliation(s)
- Karima Alim
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Alain Lescoat
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Elodie Jouan
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Valérie Lecureur
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Marc Le Vée
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Olivier Fardel
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
27
|
Magdy T, Burridge PW. Use of hiPSC to explicate genomic predisposition to anthracycline-induced cardiotoxicity. Pharmacogenomics 2021; 22:41-54. [PMID: 33448871 PMCID: PMC7923254 DOI: 10.2217/pgs-2020-0104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
The anticancer agents of the anthracycline family are commonly associated with the potential to cause severe toxicity to the heart. To solve the question of why particular a patient is predisposed to anthracycline-induced cardiotoxicity (AIC), researchers have conducted numerous pharmacogenomic studies and identified more than 60 loci associated with AIC. To date, none of these identified loci have been developed into US FDA-approved biomarkers for use in routine clinical practice. With advances in the application of human-induced pluripotent stem cell-derived cardiomyocytes, sequencing technologies and genomic editing techniques, variants associated with AIC can now be validated in a human model. Here, we provide a comprehensive overview of known genetic variants associated with AIC from the perspective of how human-induced pluripotent stem cell-derived cardiomyocytes can be used to help better explain the genomic predilection to AIC.
Collapse
Affiliation(s)
- Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
28
|
Döring H, Kreutzer D, Ritter C, Hilgeroth A. Discovery of Novel Symmetrical 1,4-Dihydropyridines as Inhibitors of Multidrug-Resistant Protein (MRP4) Efflux Pump for Anticancer Therapy. Molecules 2020; 26:molecules26010018. [PMID: 33375210 PMCID: PMC7793087 DOI: 10.3390/molecules26010018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 11/16/2022] Open
Abstract
Despite the development of targeted therapies in cancer, the problem of multidrug resistance (MDR) is still unsolved. Most patients with metastatic cancer die from MDR. Transmembrane efflux pumps as the main cause of MDR have been addressed by developed inhibitors, but early inhibitors of the most prominent and longest known efflux pump P-glycoprotein (P-gp) were disappointing. Those inhibitors have been used without knowledge about the expression of P-gp by the treated tumor. Therefore the use of inhibitors of transmembrane efflux pumps in clinical settings is reconsidered as a promising strategy in the case of the respective efflux pump expression. We discovered novel symmetric inhibitors of the symmetric efflux pump MRP4 encoded by the ABCC4 gene. MRP4 is involved in many kinds of cancer with resistance to anticancer drugs. All compounds showed better activities than the best known MRP4 inhibitor MK571 in an MRP4-overexpressing cell line assay, and the activities could be related to the various substitution patterns of aromatic residues within the symmetric molecular framework. One of the best compounds was demonstrated to overcome the MRP4-mediated resistance in the cell line model to restore the anticancer drug sensitivity as a proof of concept.
Collapse
Affiliation(s)
- Henry Döring
- Research Group of Drug Development, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (H.D.); (D.K.)
| | - David Kreutzer
- Research Group of Drug Development, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (H.D.); (D.K.)
| | - Christoph Ritter
- Department of Clinical Pharmacy, Institute of Pharmacy, Ernst Moritz Arndt University Greifswald, 17489 Greifswald, Germany;
| | - Andreas Hilgeroth
- Research Group of Drug Development, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (H.D.); (D.K.)
- Correspondence: ; Tel.: +49-345-55-25168
| |
Collapse
|
29
|
Gu ZT, Li ZZ, Wang CF. Research advances of intracellular mechanisms underlying gemcitabine resistance in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2020; 28:1150-1161. [DOI: 10.11569/wcjd.v28.i22.1150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most deadly malignant tumors that endanger human health, and pancreatic ductal adenocarcinoma (PDAC) is the most common histological type. Due to the lack of specific clinical symptoms, physical signs, and effective screening biomarkers for early stage PDAC, only 15%-20% of patients are qualified for surgical resection. Consequently, gemcitabine (GEM)-based monotherapy or combination therapy is still the most important or even the only treatment option. However, the overall response rate of PDAC to GEM is less than 20%, and GEM resistance is one of the most important factors affecting the efficacy of chemotherapy. At present, the mechanism of GEM resistance has not been clarified, which may involve congenital and acquired regulation. The heterogeneity of PDAC further increases its complexity. However, regulation of intracellular signaling pathways is the ultimate event to induce GEM resistance. This article will review the recent advances in research of GEM metabolism and regulation of signaling pathways in PDAC cells, and discuss potential GEM chemosensitization strategies, in order to improve the effective rate of chemotherapy and the outcome.
Collapse
Affiliation(s)
- Zong-Ting Gu
- Cheng-Feng Wang, State Key Lab of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zong-Ze Li
- Cheng-Feng Wang, State Key Lab of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | | |
Collapse
|
30
|
Gholamian Dehkordi N, Mirzaei SA, Elahian F. Pharmacodynamic mechanisms of anti-inflammatory drugs on the chemosensitization of multidrug-resistant cancers and the pharmacogenetics effectiveness. Inflammopharmacology 2020; 29:49-74. [PMID: 33070257 DOI: 10.1007/s10787-020-00765-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/27/2020] [Indexed: 01/07/2023]
Abstract
Drug resistance as a remarkable issue in cancer treatment is associated with inflammation which occurs through complex chemical reactions in the tumor microenvironment. Recent studies have implicated that glucocorticoids and NSAIDs are mainly useful combinations for inflammatory response modulation in chemotherapeutic protocols for cancer treatment. Immunosuppressive actions of glucocorticoids and NSAIDs are mainly mediated by the transrepression or activation regulation of inflammatory genes with different DNA-bound transcription factors including AP-1, NFAT, NF-κB, STAT and also, varying functions of COX enzymes in cancer cells. Interestingly, many investigations have proved the benefits of these anti-inflammatory agents in the quenching of multidrug resistance pathways. Numerous analyses on the ABC transporter promoters showed conserved nucleotide sequences with several DNA response elements that participate in transcriptional regulation. Furthermore, genetic variations in nucleotide sequences of membrane transporters were strongly associated with changes in these transporters' expression or function and a substantial impact on systemic drug exposure and toxicity. It appeared that several polymorphisms in MDR transporter genes especially MDR1 have influenced the regulatory mechanisms and explained differences in glucocorticoid responses.
Collapse
Affiliation(s)
- Neda Gholamian Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Abbas Mirzaei
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
31
|
Llaguno-Munive M, León-Zetina S, Vazquez-Lopez I, Ramos-Godinez MDP, Medina LA, Garcia-Lopez P. Mifepristone as a Potential Therapy to Reduce Angiogenesis and P-Glycoprotein Associated With Glioblastoma Resistance to Temozolomide. Front Oncol 2020; 10:581814. [PMID: 33123485 PMCID: PMC7571516 DOI: 10.3389/fonc.2020.581814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma, the most common primary central nervous system tumor, is characterized by extensive vascular neoformation and an area of necrosis generated by rapid proliferation. The standard treatment for this type of tumor is surgery followed by chemotherapy based on temozolomide and radiotherapy, resulting in poor patient survival. Glioblastoma is known for strong resistance to treatment, frequent recurrence and rapid progression. The aim of this study was to evaluate whether mifepristone, an antihormonal agent, can enhance the effect of temozolomide on C6 glioma cells orthotopically implanted in Wistar rats. The levels of the vascular endothelial growth factor (VEGF), and P-glycoprotein (P-gp) were examined, the former a promoter of angiogenesis that facilitates proliferation, and the latter an efflux pump transporter linked to drug resistance. After a 3-week treatment, the mifepristone/temozolomide regimen had decreased the level of VEGF and P-gp and significantly reduced tumor proliferation (detected by PET/CT images based on 18F-fluorothymidine uptake). Additionally, mifepristone proved to increase the intracerebral concentration of temozolomide. The lower level of O6-methylguanine-DNA-methyltransferase (MGMT) (related to DNA repair in tumors) previously reported for this combined treatment was herein confirmed. After the mifepristone/temozolomide treatment ended, however, the values of VEGF, P-gp, and MGMT increased and reached control levels by 14 weeks post-treatment. There was also tumor recurrence, as occurred when administering temozolomide alone. On the other hand, temozolomide led to 100% mortality within 26 days after beginning the drug treatment, while mifepristone/temozolomide enabled 70% survival 60–70 days and 30% survived over 100 days, suggesting that mifepristone could possibly act as a chemo-sensitizing agent for temozolomide.
Collapse
Affiliation(s)
- Monserrat Llaguno-Munive
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico.,Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sebastián León-Zetina
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Inés Vazquez-Lopez
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Luis A Medina
- Unidad de Investigación Biomédica en Cáncer INCan-UNAM, Instituto Nacional de Cancerología, Mexico City, Mexico.,Instituto de Física, Universidad Nacional Autónoma de México, Coyoacán, Mexico City, Mexico
| | - Patricia Garcia-Lopez
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
32
|
Ledezma-Gallegos F, Jurado R, Mir R, Medina LA, Mondragon-Fuentes L, Garcia-Lopez P. Liposomes Co-Encapsulating Cisplatin/Mifepristone Improve the Effect on Cervical Cancer: In Vitro and In Vivo Assessment. Pharmaceutics 2020; 12:E897. [PMID: 32971785 PMCID: PMC7558205 DOI: 10.3390/pharmaceutics12090897] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 01/24/2023] Open
Abstract
Cervical cancer is usually diagnosed in the later stages despite many campaigns for early detection and continues to be a major public health problem. The standard treatment is cisplatin-based chemotherapy plus radiotherapy, but patient response is far from ideal. In the research for new drugs that enhance the activity of cisplatin, different therapeutic agents have been tested, among them the antiprogestin mifepristone. Nevertheless, the efficacy of cisplatin is limited by its low specificity for tumor tissue, which causes severe side effects. Additionally, cervical tumors often become drug resistant. These problems could possibly be addressed by the use of liposome nanoparticles to encapsulate drugs and deliver them to the target. The aim of this study was to prepare liposome nanoparticles that co-encapsulate cisplatin and mifepristone, evaluate their cytotoxicity against HeLa cells and in vivo with subcutaneous inoculations of xenografts in nu/nu mice, and examine some plausible mechanisms of action. The liposomes were elaborated by the reverse-phase method and characterized by physicochemical tests. The nanoparticles had a mean particle size of 109 ± 5.4 nm and a Zeta potential of -38.7 ± 1.2 mV, the latter parameter indicating a stable formulation. These drug-loaded liposomes significantly decreased cell viability in vitro and tumor size in vivo, without generating systemic toxicity in the animals. There was evidence of cell cycle arrest and increased apoptosis. The promising results with the co-encapsulation of cisplatin/mifepristone warrant further research.
Collapse
Affiliation(s)
- Fabricio Ledezma-Gallegos
- Laboratorio de Farmacologia, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd. México 14080, Mexico; (F.L.-G.); (R.J.); (R.M.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Coyacán, Cd. México 04510, Mexico
| | - Rafael Jurado
- Laboratorio de Farmacologia, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd. México 14080, Mexico; (F.L.-G.); (R.J.); (R.M.)
| | - Roser Mir
- Laboratorio de Farmacologia, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd. México 14080, Mexico; (F.L.-G.); (R.J.); (R.M.)
| | - Luis Alberto Medina
- Unidad de Investigación Biomédica en Cáncer INCan-UNAM, Instituto Nacional de Cancerología, Cd. México 14080, Mexico; (L.A.M); (L.M.-F.)
- Instituto de Física, Universidad Nacional Autónoma de México, Coyoacán, Cd. México 04510, Mexico
| | - Laura Mondragon-Fuentes
- Unidad de Investigación Biomédica en Cáncer INCan-UNAM, Instituto Nacional de Cancerología, Cd. México 14080, Mexico; (L.A.M); (L.M.-F.)
| | - Patricia Garcia-Lopez
- Laboratorio de Farmacologia, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd. México 14080, Mexico; (F.L.-G.); (R.J.); (R.M.)
| |
Collapse
|
33
|
Kreutzer D, Ritter CA, Hilgeroth A. Novel Nonsymmetrical 1,4-Dihydropyridines as Inhibitors of Nonsymmetrical MRP-Efflux Pumps for Anticancer Therapy. Pharmaceuticals (Basel) 2020; 13:ph13070146. [PMID: 32660005 PMCID: PMC7407134 DOI: 10.3390/ph13070146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 11/22/2022] Open
Abstract
Cancer is a strong global burden with increasing numbers of diseases and ongoing anticancer drug resistance. The number of structurally novel anticancer drugs is strongly limited. They cause high costs for the social health systems. Most critical so-called multidrug resistances (MDR) are caused by transmembrane efflux pumps that transport drugs with various structures out of the cancer cells. Multidrug resistance proteins (MRPs) type 1 and 2 are found overexpressed in various kinds of cancer. There is a strong need for inhibitors of those efflux pumps. We developed novel nonsymmetrical 1,4-dihydropyridines as novel inhibitors of cancer relevant MRP types 1 and 2. The structure-dependent activities of the differently substituted derivatives were evaluated in cellular assays of respective cancer cells and are discussed. Promising candidates were identified. One candidate was demonstrated to resensitize a cisplatin resistant cancer cell line and thus to overcome the anticancer drug resistance.
Collapse
Affiliation(s)
- David Kreutzer
- Institute of Pharmacy, Research Group of Drug Development, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany;
| | - Christoph A. Ritter
- Institute of Pharmacy, Department of Clinical Pharmacy, University of Greifswald, 17489 Greifswald, Germany;
| | - Andreas Hilgeroth
- Institute of Pharmacy, Research Group of Drug Development, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany;
- Correspondence: ; Tel.: +49-345-55-25168
| |
Collapse
|
34
|
Mello FV, de Moraes GN, Maia RC, Kyeremateng J, Iram SH, Santos-Oliveira R. The Effect of Nanosystems on ATP-Binding Cassette Transporters: Understanding the Influence of Nanosystems on Multidrug Resistance Protein-1 and P-glycoprotein. Int J Mol Sci 2020; 21:E2630. [PMID: 32290047 PMCID: PMC7178121 DOI: 10.3390/ijms21072630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
The cancer multidrug resistance is involved in the failure of several treatments during cancer treatment. It is a phenomenon that has been receiving great attention in the last years due to the sheer amount of mechanisms discovered and involved in the process of resistance which hinders the effectiveness of many anti-cancer drugs. Among the mechanisms involved in the multidrug resistance, the participation of ATP-binding cassette (ABC) transporters is the main one. The ABC transporters are a group of plasma membrane and intracellular organelle proteins involved in the process of externalization of substrates from cells, which are expressed in cancer. They are involved in the clearance of intracellular metabolites as ions, hormones, lipids and other small molecules from the cell, affecting directly and indirectly drug absorption, distribution, metabolism and excretion. Other mechanisms responsible for resistance are the signaling pathways and the anti- and pro-apoptotic proteins involved in cell death by apoptosis. In this study we evaluated the influence of three nanosystem (Graphene Quantum Dots (GQDs), mesoporous silica (MSN) and poly-lactic nanoparticles (PLA)) in the main mechanism related to the cancer multidrug resistance such as the Multidrug Resistance Protein-1 and P-glycoprotein. We also evaluated this influence in a group of proteins involved in the apoptosis-related resistance including cIAP-1, XIAP, Bcl-2, BAK and Survivin proteins. Last, colonogenic and MTT (3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium bromide) assays have also been performed. The results showed, regardless of the concentration used, GQDs, MSN and PLA were not cytotoxic to MDA-MB-231 cells and showed no impairment in the colony formation capacity. In addition, it has been observed that P-gp membrane expression was not significantly altered by any of the three nanomaterials. The results suggest that GQDs nanoparticles would be suitable for the delivery of other multidrug resistance protein 1 (MRP1) substrate drugs that bind to the transporter at the same binding pocket, while MSN can strongly inhibit doxorubicin efflux by MRP1. On the other hand, PLA showed moderate inhibition of doxorubicin efflux by MRP1 suggesting that this nanomaterial can also be useful to treat MDR (Multidrug resistance) due to MRP1 overexpression.
Collapse
Affiliation(s)
- Francisco V.C. Mello
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rua Helio de Almeida 75, Ilha do Fundão, CEP 21941-614 Rio de Janeiro, Brazil;
| | - Gabriela N. de Moraes
- Laboratory of Cellular and Molecular Hemato-Oncology, Program of Molecular Hemato-Oncology, Brazilian National Cancer Institute (INCA), CEP 20230130 Rio de Janeiro, Brazil; (G.N.d.M.); (R.C.M.)
| | - Raquel C. Maia
- Laboratory of Cellular and Molecular Hemato-Oncology, Program of Molecular Hemato-Oncology, Brazilian National Cancer Institute (INCA), CEP 20230130 Rio de Janeiro, Brazil; (G.N.d.M.); (R.C.M.)
| | - Jennifer Kyeremateng
- Department of Chemistry & Biochemistry, College of Natural Sciences, South Dakota State University, Brookings, SD 57007, USA; (J.K.); (S.H.I.)
| | - Surtaj Hussain Iram
- Department of Chemistry & Biochemistry, College of Natural Sciences, South Dakota State University, Brookings, SD 57007, USA; (J.K.); (S.H.I.)
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rua Helio de Almeida 75, Ilha do Fundão, CEP 21941-614 Rio de Janeiro, Brazil;
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Zona Oeste State University, Campo Grande, CEP 23070200 Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Huisman BW, Burggraaf J, Vahrmeijer AL, Schoones JW, Rissmann RA, Sier CFM, van Poelgeest MIE. Potential targets for tumor-specific imaging of vulvar squamous cell carcinoma: A systematic review of candidate biomarkers. Gynecol Oncol 2020; 156:734-743. [PMID: 31928804 DOI: 10.1016/j.ygyno.2019.12.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Vulvar squamous cell carcinoma (VSCC) is a rare malignancy with an increasing incidence, especially in young women. Surgical treatment of VSCC is associated with significant morbidity and high recurrence rates, which is related to the limited ability to distinguish (pre)malignant from healthy tissue. There is a need for new tools for specific real-time detection of occult tumor lesions and localization of cancer margins in patients with VSCC. Several tumor-specific imaging techniques are developed to recognize malignant tissue by targeting tumor markers. We present a systematic review to identify, evaluate, and summarize potential markers for tumor-specific imaging of VSCC. METHODS Relevant papers were identified by a systematic cross-database literature search developed with assistance of an experienced librarian. Data were extracted from eligible papers and reported based on the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines. VSCC-specific tumor markers were valued based on a weighted scoring system, in which each biomarker was granted points based on ranked eligibility criteria: I) percentage expression, II) sample size, and III) in vivo application. RESULTS In total 627 papers were included of which 22 articles met the eligibility criteria. Twelve VSCC-specific tumor markers were identified and of these 7 biomarkers were considered most promising: EGFR, CD44v6, GLUT1, MRP1, MUC1, CXCR-4 and VEGF-A. DISCUSSION This overview identified 7 potential biomarkers that can be used in the development of VSCC-specific tracers for real-time and precise localization of tumor tissue before, during, and after treatment. These biomarkers were identified in a small number of samples, without discriminating for VSCC-specific hallmarks such as HPV-status. Before clinical development, experimental studies should first aim at validation of these biomarkers using immunohistochemistry and cell line-based examination, discriminating for HPV-status and the expression rate in lymph nodes and precursor lesions.
Collapse
Affiliation(s)
- B W Huisman
- Centre for Human Drug Research, Zernikedreef 8, 2333CL Leiden, the Netherlands; Department of Gynecology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands.
| | - J Burggraaf
- Centre for Human Drug Research, Zernikedreef 8, 2333CL Leiden, the Netherlands; Leiden Academic Center for Drug Research, Leiden University, Einstein weg 55, 2333 CC Leiden, the Netherlands; Department of Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands.
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands.
| | - J W Schoones
- Walaeus Library, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC, the Netherlands.
| | - R A Rissmann
- Centre for Human Drug Research, Zernikedreef 8, 2333CL Leiden, the Netherlands; Leiden Academic Center for Drug Research, Leiden University, Einstein weg 55, 2333 CC Leiden, the Netherlands.
| | - C F M Sier
- Department of Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands.
| | - M I E van Poelgeest
- Centre for Human Drug Research, Zernikedreef 8, 2333CL Leiden, the Netherlands; Department of Gynecology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands.
| |
Collapse
|