1
|
Nathani A, Sun L, Li Y, Lazarte J, Aare M, Singh M. Targeting EGFR-TKI resistance in lung cancer: Role of miR-5193/miR-149-5p loaded NK-EVs and Carboplatin combination. Int J Pharm 2025; 675:125573. [PMID: 40204039 DOI: 10.1016/j.ijpharm.2025.125573] [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: 02/02/2025] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Lung cancer remains the leading cause of cancer-related deaths, and there is an urgent need for innovative therapies. MicroRNA (miRNA)-based gene therapy has shown promise, but efficient delivery systems are required for its success. This study investigates the use of extracellular vehicles (EVs) secreted by natural killer (NK) cells as delivery systems for miRNAs targeting PD-L1/PD-1 immune checkpoint and FOXM1, in combination with Carboplatin, to enhance anticancer efficacy in lung cancer models. NK-EVs were isolated from NK92-MI cells and characterized using nanoparticle tracking analysis (NTA), proteomics and Western blotting, confirming their exosomal characteristics. Gene ontology profiling and RNA-seq identified highly expressed miRNAs such as miR-5193 and miR-149-5p, which were loaded into NK-EVs via electroporation. Agarose gel electrophoresis confirmed their entrapment and Quickdrop spectrophotometer was used to estimate the quantity. In vitro, miRNA-loaded NK-EVs demonstrated significant cytotoxicity against Osimertinib-resistant PDX (TM0019, Jackson Labs) and H1975R (with L858R mutations) lung cancer cells, with approximately 1.2 to 1.6-fold (p < 0.01) decrease in cell viability compared to NK-EVs alone. In vivo, the combination of miRNA-loaded NK-EVs and Carboplatin significantly reduced tumor volumes (3.5 to 4-fold, p < 0.001) in PDX and H1975R xenograft models, with the most pronounced effect observed in combination therapies. Western blot analysis showed downregulation of tumor-associated markers: PD-1/PD-L1, FOXM1, Survivin, NF-κB and others vs untreated group, p < 0.001) suggesting immune checkpoint inhibition, apoptosis and anti-inflammatory activity. These findings highlight the potential of NK-EVs as effective carriers for miRNAs in combination with chemotherapy, offering a promising therapeutic strategy for NSCLC with EGFR mutations.
Collapse
Affiliation(s)
- Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA; Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Jassy Lazarte
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA.
| |
Collapse
|
2
|
Liu X, Feng C, Yan L, Cao J, Zhu X, Li M, Zhao G. Calcium channels as pharmacological targets for cancer therapy. Clin Exp Med 2025; 25:94. [PMID: 40131496 PMCID: PMC11937194 DOI: 10.1007/s10238-025-01632-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Ca2+, as critical second messengers in biological processes, plays a pivotal role in the regulation of diverse cellular signaling pathways. The dysregulation of calcium signaling is intricately linked to the progression of various cancers. The capacity of Ca2+ to modulate cell death and proliferation, along with its potential for pharmacological manipulation, presents a promising avenue for the development of novel cancer therapeutics. This review provides a comprehensive overview of the classification of Ca2+ channels and their mechanisms of action in oncogenesis, explores the application of Ca2+ blockers in cancer treatment, and underscores the importance of conducting further clinical trials.
Collapse
Affiliation(s)
- Xiaozhen Liu
- Department of Medical and Radiation Oncology, Linyi People's Hospital, Linyi, 276000, China
| | - Changyun Feng
- Department of Pediatrics, Linyi Maternal and Child Health Hospital, Linyi, 276000, China
| | - Li Yan
- Department of Medical and Radiation Oncology, Linyi People's Hospital, Linyi, 276000, China
| | - Jili Cao
- Zhejiang Key Laboratory of Disease-Syndrome Integration for Cancer Prevention and Treatment, Tongde Hospital of Zhejiang Province Afflicted to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, 310012, Zhejiang, China
| | - Xinping Zhu
- Zhejiang Key Laboratory of Disease-Syndrome Integration for Cancer Prevention and Treatment, Tongde Hospital of Zhejiang Province Afflicted to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, 310012, Zhejiang, China
| | - Mingqian Li
- Zhejiang Key Laboratory of Disease-Syndrome Integration for Cancer Prevention and Treatment, Tongde Hospital of Zhejiang Province Afflicted to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, 310012, Zhejiang, China.
| | - Guizhi Zhao
- The Integrated Traditional Chinese and Western Medicine School of Clinical Medicne (Tongde Hospital of Zhejiang Province), Zhejiang Chinese Medical University, Hangzhou, 310012, Zhejiang, China.
| |
Collapse
|
3
|
Koyama S, Etkins J, Jun J, Miller M, So GC, Gisch DL, Eadon MT. Utilization of Cannabidiol in Post-Organ-Transplant Care. Int J Mol Sci 2025; 26:699. [PMID: 39859413 PMCID: PMC11765766 DOI: 10.3390/ijms26020699] [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: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Cannabidiol (CBD) is one of the major phytochemical constituents of cannabis, Cannabis sativa, widely recognized for its therapeutic potential. While cannabis has been utilized for medicinal purposes since ancient times, its psychoactive and addictive properties led to its prohibition in 1937, with only the medical use being reauthorized in 1998. Unlike tetrahydrocannabinol (THC), CBD lacks psychoactive and addictive properties, yet the name that suggests its association with cannabis has significantly contributed to its public visibility. CBD exhibits diverse pharmacological properties, most notably anti-inflammatory effects. Additionally, it interacts with key drug-metabolizing enzyme families, including cytochrome P450 (CYP) and uridine 5'-diphospho-glucuronosyltransferase (UGT), which mediate phase I and phase II metabolism, respectively. By binding to these enzymes, CBD can inhibit the metabolism of co-administered drugs, which can potentially enhance their toxicity or therapeutic effects. Mild to moderate adverse events associated with CBD use have been reported. Advances in chemical formulation techniques have recently enabled strategies to minimize these effects. This review provides an overview of CBD, covering its historical background, recent clinical trials, adverse event profiles, and interactions with molecular targets such as receptors, channels, and enzymes. We particularly emphasize the mechanisms underlying its anti-inflammatory effects and interaction with drugs relevant to organ transplantation. Finally, we explore recent progress in the chemical formulation of CBD in order to enhance its bioavailability, which will enable decreasing the dose to use and increase its safety and efficacy.
Collapse
Affiliation(s)
- Sachiko Koyama
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Jumar Etkins
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Joshua Jun
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Matthew Miller
- College of Human Ecology, Cornell University, Ithaca, NY 14850, USA;
| | - Gerald C. So
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Debora L. Gisch
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| | - Michael T. Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.E.); (J.J.); (D.L.G.); (M.T.E.)
| |
Collapse
|
4
|
DaCunza JT, Wickman JR, Ajit SK. miRNA packaging into small extracellular vesicles and implications in pain. Pain Rep 2024; 9:e1198. [PMID: 39450410 PMCID: PMC11500789 DOI: 10.1097/pr9.0000000000001198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/17/2024] [Accepted: 06/30/2024] [Indexed: 10/26/2024] Open
Abstract
Extracellular vesicles (EVs) are a heterogenous group of lipid bilayer bound particles naturally released by cells. These vesicles are classified based on their biogenesis pathway and diameter. The overlap in size of exosomes generated from the exosomal pathway and macrovesicles that are pinched off from the surface of the plasma membrane makes it challenging to isolate pure populations. Hence, isolated vesicles that are less than 200 nm are called small extracellular vesicles (sEVs). Extracellular vesicles transport a variety of cargo molecules, and multiple mechanisms govern the packaging of cargo into sEVs. Here, we discuss the current understanding of how miRNAs are targeted into sEVs, including the role of RNA binding proteins and EXOmotif sequences present in miRNAs in sEV loading. Several studies in human pain disorders and rodent models of pain have reported alterations in sEV cargo, including miRNAs. The sorting mechanisms and target regulation of miR-939, a miRNA altered in individuals with complex regional pain syndrome, is discussed in the context of inflammation. We also provide a broad overview of the therapeutic strategies being pursued to utilize sEVs in the clinic and the work needed to further our understanding of EVs to successfully deploy sEVs as a pain therapeutic.
Collapse
Affiliation(s)
- Jason T. DaCunza
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
- Molecular & Cell Biology & Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jason R. Wickman
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Seena K. Ajit
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
5
|
Liu R, Zou Z, Zhang Z, He H, Xi M, Liang Y, Ye J, Dai Q, Wu Y, Tan H, Zhong W, Wang Z, Liang Y. Evaluation of glucocorticoid-related genes reveals GPD1 as a therapeutic target and regulator of sphingosine 1-phosphate metabolism in CRPC. Cancer Lett 2024; 605:217286. [PMID: 39413958 DOI: 10.1016/j.canlet.2024.217286] [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: 09/21/2023] [Revised: 09/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Prostate cancer (PCa) is an androgen-dependent disease, with castration-resistant prostate cancer (CRPC) being an advanced stage that no longer responds to androgen deprivation therapy (ADT). Mounting evidence suggests that glucocorticoid receptors (GR) confer resistance to ADT in CRPC patients by bypassing androgen receptor (AR) blockade. GR, as a novel therapeutic target in CRPC, has attracted substantial attention worldwide. This study utilized bioinformatic analysis of publicly available CRPC single-cell data to develop a consensus glucocorticoid-related signature (Glu-sig) that can serve as an independent predictor for relapse-free survival. Our results revealed that the signature demonstrated consistent and robust performance across seven publicly accessible datasets and an internal cohort. Furthermore, our findings demonstrated that glycerol-3-phosphate dehydrogenase 1 (GPD1) in Glu-sig can significantly promote CRPC progression by mediating the cell cycle pathway. Additionally, GPD1 was shown to be regulated by GR, with the GR antagonist mifepristone enhancing the anti-tumorigenic effects of GPD1 in CRPC cells. Mechanistically, targeting GPD1 induced the production of sphingosine 1-phosphate (S1P) and enhanced histone acetylation, thereby inducing the transcription of p21 that involved in cell cycle regulation. In conclusion, Glu-sig could serve as a robust and promising tool to improve the clinical outcomes of PCa patients, and modulating the GR/GPD1 axis that promotes tumor growth may be a promising approach for delaying CRPC progression.
Collapse
Affiliation(s)
- Ren Liu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihao Zou
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Laboratory, Guangzhou, China
| | - Zhengrong Zhang
- Department of Urology, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Huichan He
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Ming Xi
- Department of Urology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Yingke Liang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qishan Dai
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongding Wu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huijing Tan
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weide Zhong
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Laboratory, Guangzhou, China; Macau Institute of Systems Engineering, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China.
| | - Zongren Wang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yuxiang Liang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Palakurthi SS, Shah B, Kapre S, Charbe N, Immanuel S, Pasham S, Thalla M, Jain A, Palakurthi S. A comprehensive review of challenges and advances in exosome-based drug delivery systems. NANOSCALE ADVANCES 2024; 6:5803-5826. [PMID: 39484149 PMCID: PMC11523810 DOI: 10.1039/d4na00501e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/22/2024] [Indexed: 11/03/2024]
Abstract
Exosomes or so-called natural nanoparticles have recently shown enormous potential for targeted drug delivery systems. Several studies have reported that exosomes as advanced drug delivery platforms offer efficient targeting of chemotherapeutics compared to individual polymeric nanoparticles or liposomes. Taking structural constituents of exosomes, viz., proteins, nucleic acids, and lipids, into consideration, exosomes are the most promising carriers as genetic messengers and for treating genetic deficiencies or tumor progression. Unfortunately, very little attention has been paid to the factors like source, scalability, stability, and validation that contribute to the quality attributes of exosome-based drug products. Some studies suggested that exosomes were stable at around -80 °C, which is impractical for storing pharmaceutical products. Currently, no reports on the shelf-life and in vivo stability of exosome formulations are available. Exosomes are quickly cleared from blood circulation, and their in vivo distribution depends on the source. Considering these challenges, further studies are necessary to address major limitations such as poor drug loading, reduced in vivo stability, a need for robust, economical, and scalable production methods, etc., which may unlock the potential of exosomes in clinical applications. A few reports based on hybrid exosomes involving hybridization between different cell/tumor/macrophage-derived exosomes with synthetic liposomes through membrane fusion have shown to overcome some limitations associated with natural or synthetic exosomes. Yet, sufficient evidence is indispensable to prove their stability and clinical efficacy.
Collapse
Affiliation(s)
- Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Brijesh Shah
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Sumedha Kapre
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Nitin Charbe
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Susan Immanuel
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Sindhura Pasham
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Maharshi Thalla
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Ankit Jain
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| |
Collapse
|
7
|
Nathani A, Aare M, Sun L, Bagde A, Li Y, Rishi A, Singh M. Unlocking the Potential of Camel Milk-Derived Exosomes as Novel Delivery Systems: Enhanced Bioavailability of ARV-825 PROTAC for Cancer Therapy. Pharmaceutics 2024; 16:1070. [PMID: 39204415 PMCID: PMC11359469 DOI: 10.3390/pharmaceutics16081070] [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: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
This study investigates the use of camel milk-derived exosomes (CMEs) as carriers for ARV-825, an anticancer agent targeting bromodomain-containing protein 4 (BRD4), in oral chemotherapy. CMEs were isolated and characterized, and ARV-825-loaded CME formulations were prepared and evaluated through various in vitro and in vivo tests. The ARV-825-CME formulation exhibited an entrapment efficiency of 42.75 ± 5.05%, a particle size of 136.8 ± 1.94 nm, and a zeta potential of -32.75 ± 0.70 mV, ensuring stability and sustained drug release. In vitro studies showed a 5.4-fold enhancement in drug release kinetics compared to the free ARV-825 solution. Permeability studies indicated a 3.2-fold increase in apparent permeability, suggesting improved cellular uptake. Cytotoxicity assays demonstrated potent anticancer activity, with IC50 values decreasing by 1.5 to 2-fold in cancer cell lines SF8628 DIPG and H1975R (resistant to Osimertinib). In vivo pharmacokinetic studies in Sprague-Dawley rats revealed superior systemic absorption and bioavailability of ARV-825 from CMEs, with a 2.55-fold increase in plasma concentration and a 5.56-fold increase in AUC. Distribution studies confirmed absorption through the ileum. This research highlights the potential of CMEs as a promising delivery platform for ARV-825, enhancing its therapeutic efficacy and offering a novel approach to cancer treatment.
Collapse
Affiliation(s)
- Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA; (L.S.); (Y.L.)
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA; (L.S.); (Y.L.)
| | - Arun Rishi
- Department of Oncology, John D. Dingell VA Medical Center, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| |
Collapse
|
8
|
Kaur S, Mendonca P, Soliman KFA. The Anticancer Effects and Therapeutic Potential of Kaempferol in Triple-Negative Breast Cancer. Nutrients 2024; 16:2392. [PMID: 39125273 PMCID: PMC11314279 DOI: 10.3390/nu16152392] [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: 06/15/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Breast cancer is the second-leading cause of cancer death among women in the United States. Triple-negative breast cancer (TNBC), a subtype of breast cancer, is an aggressive phenotype that lacks estrogen (ER), progesterone (PR), and human epidermal growth (HER-2) receptors, which is challenging to treat with standardized hormonal therapy. Kaempferol is a natural flavonoid with antioxidant, anti-inflammatory, neuroprotective, and anticancer effects. Besides anti-tumorigenic, antiproliferative, and apoptotic effects, kaempferol protects non-cancerous cells. Kaempferol showed anti-breast cancer effects by inducing DNA damage and increasing caspase 3, caspase 9, and pAMT expression, modifying ROS production by Nrf2 modulation, inducing apoptosis by increasing cleaved PARP and Bax and downregulating Bcl-2 expression, inducing cell cycle arrest at the G2/M phase; inhibiting immune evasion by modulating the JAK-STAT3 pathway; and inhibiting the angiogenic and metastatic potential of tumors by downregulating MMP-3 and MMP-9 levels. Kaempferol holds promise for boosting the efficacy of anticancer agents, complementing their effects, or reversing developed chemoresistance. Exploring novel TNBC molecular targets with kaempferol could elucidate its mechanisms and identify strategies to overcome limitations for clinical application. This review summarizes the latest research on kaempferol's potential as an anti-TNBC agent, highlighting promising but underexplored molecular pathways and delivery challenges that warrant further investigation to achieve successful clinical translation.
Collapse
Affiliation(s)
- Sukhmandeep Kaur
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA;
| |
Collapse
|
9
|
Nathani A, Sun L, Khan I, Aare M, Bagde A, Li Y, Singh M. Combined Role of Interleukin-15 Stimulated Natural Killer Cell-Derived Extracellular Vesicles and Carboplatin in Osimertinib-Resistant H1975 Lung Cancer Cells with EGFR Mutations. Pharmaceutics 2024; 16:83. [PMID: 38258094 PMCID: PMC10821370 DOI: 10.3390/pharmaceutics16010083] [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: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
In this study, we evaluated IL-15 stimulated natural killer cell-derived EVs (NK-EVs) as therapeutic agents in vitro and in vivo in Osimertinib-resistant lung cancer (H1975R) with EGFR mutations (L858R) in combination with carboplatin (CBP). NK-EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis, and atomic force microscopy imaging revealed vesicles with a spherical form and sizes meeting the criteria of exosomal EVs. Further, Western blot studies demonstrated the presence of regular EV markers along with specific NK markers (perforin and granzyme). EVs were also characterized by proteomic analysis, which demonstrated that EVs had proteins for natural killer cell-mediated cytotoxicity (Granzyme B) and T cell activation (perforin and plastin-2). Gene oncology analysis showed that these differentially expressed proteins are involved in programmed cell death and positive regulation of cell death. Further, isolated NK-EVs were cytotoxic to H1975R cells in vitro in 2D and 3D cell cultures. CBP's IC50 was reduced by approximately in 2D and 3D cell cultures when combined with NK-EVs. The EVs were then combined with CBP and administered by i.p. route to H1975R tumor xenografts, and a significant reduction in tumor volume in vivo was observed. Our findings show for the first time that NK-EVs target the PD-L1/PD-1 immunological checkpoint to induce apoptosis and anti-inflammatory response by downregulation of SOD2, PARP, BCL2, SET, NF-κB, and TGF-ß. The ability to isolate functional NK-EVs on a large scale and use them with platinum-based drugs may lead to new clinical applications. The results of the present study suggest the possibility of the combination of NK-cell-derived EVs and CBP as a viable immunochemotherapeutic strategy for resistant cancers.
Collapse
Affiliation(s)
- Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Islauddin Khan
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| |
Collapse
|
10
|
Li X, Jing Z, Li X, Liu L, Xiao X, Zhong Y, Ren Z. The role of exosomes in cancer-related programmed cell death. Immunol Rev 2024; 321:169-180. [PMID: 37950410 DOI: 10.1111/imr.13286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Cancer arises from the growth and division of uncontrolled erroneous cells. Programmed cell death (PCD), or regulated cell death (RCD), includes natural processes that eliminate damaged or abnormal cells. Dysregulation of PCD is a hallmark of cancer, as cancer cells often evade cell death and continue to proliferate. Exosomes nanoscale extracellular vesicles secreted by different types of cells carrying a variety of molecules, including nucleic acids, proteins, and lipids, to have indispensable role in the communication between cells, and can influence various cellular processes, including PCD. Exosomes have been shown to modulate PCD in cancer cells by transferring pro- or antideath molecules to neighboring cells. Additionally, exosomes can facilitate the spread of PCD to surrounding cancer cells, making them promising in the treatment of various cancers. The exosomes' diagnostic potential in cancer is also an active area of research. Exosomes can be isolated from a wide range of bodily fluids and tissues, such as blood and urine, and can provide a noninvasive way to monitor cancer progression and treatment response. Furthermore, exosomes have also been employed as a delivery system for therapeutic agents. By engineering exosomes to carry drugs or other therapeutic molecules, they can be targeted specifically to cancer cells, reducing toxicity to healthy tissues. Here, we discussed exosomes in the diagnosis and prevention of cancers, tumor immunotherapy, and drug delivery, as well as in different types of PCD.
Collapse
Affiliation(s)
- Xin Li
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Zuoqian Jing
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Xuejie Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Lei Liu
- Department of Ophthalmology, Guangdong provincial People's hospital, Guangzhou, China
| | - Xiang Xiao
- Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Yifan Zhong
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Zihan Ren
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Aqil F, Gupta R. Exosomes as emerging nanoplatform in cancer therapy. Cancer Lett 2023; 574:216394. [PMID: 37716464 PMCID: PMC10569016 DOI: 10.1016/j.canlet.2023.216394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023]
Affiliation(s)
- Farrukh Aqil
- Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA; Department of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| | - Ramesh Gupta
- Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA; 3P Biotechnologies, Inc., 580 S. Preston Street, Louisville, KY, 40202, USA.
| |
Collapse
|