1
|
Wu J, Liu X, Huang S, Liu W. Identification of a Cancer Stem Cell-Related Gene Signature in Hepatocellular Carcinoma Based on Single-Cell RNA-Seq and Bulk RNA-Seq Analysis. Int J Mol Sci 2025; 26:2933. [PMID: 40243557 PMCID: PMC11988464 DOI: 10.3390/ijms26072933] [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: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) are a heterogeneous group of tumor cells that play a significant role in tumorigenesis, therapeutic resistance, and recurrence in liver hepatocellular carcinoma (LIHC). This study combines clinical data sets from The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) with bulk RNA sequencing data. This study also features the GSE156625 single-cell RNA sequencing (scRNA) data set from the GEO to explore the prognostic significance of CSC biomarkers (BCSCs) in LIHC. In this research, we introduce a developed prognostic risk model that relies on nine specific BCSCs, including ADM, CCL5, CD274, DLGAP5, HOXD9, IGF1, S100A9, SOCS2, and TNFRSF11B. It was found that high-risk patients experience shorter overall survival rates when compared to low-risk patients. Additionally, the study characterized the composition of immune cells within the tumor microenvironment (TME) and revealed significant variations in gene-expression levels and mutation rates between different risk groups. The model suggests that liver cancer progression might be driven by immune evasion independent of PD-L1 and highlights the potential of the low-risk BCSC group being sensitive to various treatments. Our findings offer a promising foundation for personalized LIHC therapy and highlight the need for further experimental validation of the roles of these CSCs in disease progression.
Collapse
Affiliation(s)
- Jing Wu
- Medical School, Hubei Minzu University, Enshi 445000, China; (X.L.); (S.H.)
| | - Xu Liu
- Medical School, Hubei Minzu University, Enshi 445000, China; (X.L.); (S.H.)
| | - Sheng Huang
- Medical School, Hubei Minzu University, Enshi 445000, China; (X.L.); (S.H.)
| | - Wei Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
2
|
Kast RE. UBC4: A Repurposed Drug Regimen for Adjunctive Use During Bladder Cancer Treatment. Biomedicines 2025; 13:706. [PMID: 40149682 PMCID: PMC11940094 DOI: 10.3390/biomedicines13030706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
After it has metastasized, bladder cancer, the malignant transformation of the bladder urothelium, continues to be a common cause of death after maximal use of all currently available standard treatments. To address this problem in 2025, the drug repurposing movement within oncology aims to identify medicines in common general medical care use that have data indicating that they can interfere or inhibit a growth driving element that has been identified in bladder cancer. This paper now outlines extensive preclinical data showing that four drugs from general medical practice meet these criteria-the melatonergic drug ramelteon, the antidepressant fluoxetine, the antibiotic dapsone, and the analgesic drug celecoxib. This is the UBC4 regimen, meant as a possible adjunct added to standard treatments of metastatic bladder cancer. Three factors justify a clinical pilot trial of UBC4: (1) the UBC4 drugs are usually well tolerated and carry a low risk of harm, (2) the commonly fatal outcome of bladder cancer once it has widely metastasized, plus (3) the strong preclinical database showing UBC growth inhibition by each of the individual UBC4 drugs as outlined in this paper.
Collapse
Affiliation(s)
- Richard E Kast
- IIAIGC Study Center, 11 Arlington Ct, Burlington, VT 05408, USA
| |
Collapse
|
3
|
Kast RE. Potential Benefits of Adding Alendronate, Celecoxib, Itraconazole, Ramelteon, and Simvastatin to Endometrial Cancer Treatment: The EC5 Regimen. Curr Issues Mol Biol 2025; 47:153. [PMID: 40136407 PMCID: PMC11941490 DOI: 10.3390/cimb47030153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Metastatic endometrial cancer continues to be a common cause of death as of 2024, even after maximal use of all currently available standard treatments. To address this problem of metastatic cancer generally in 2025, the drug repurposing movement within oncology identifies medicines in common general medical use that have clinical or preclinical experimental data indicating that they interfere with or inhibit a specific growth driving element identified in a given cancer. The drug repurposing movement within oncology also uses data from large scale in vitro screens of thousands of drugs, looking for simple empirical growth inhibition in a given cancer type. This paper outlines the data showing that five drugs from general medical practice meet these evidence criteria for inhibition of endometrial cancer growth, the EC5 regimen. The EC5 regimen uses the osteoporosis treatment drug, alendronate; the analgesic drug, celecoxib; the antifungal drug, itraconazole; the sleep aid, ramelteon; and the cholesterol lowering drug, simvastatin. Side effects seen with these drugs are usually minimal and easily tolerated by patients.
Collapse
|
4
|
Li XK, Amirkhanyan Z, Grebinyk A, Gross M, Komar Y, Riemer F, Asoyan A, Boonpornprasert P, Borchert P, Davtyan H, Dmytriiev D, Frohme M, Hoffmann A, Krasilnikov M, Loisch G, Lotfi Z, Müller F, Schmitz M, Obier F, Oppelt A, Philipp S, Richard C, Vashchenko G, Villani D, Worm S, Stephan F. Demonstration of ultra-high dose rate electron irradiation at FLASH lab@PITZ. Phys Med Biol 2025; 70:055010. [PMID: 39907068 DOI: 10.1088/1361-6560/adb276] [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: 10/16/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025]
Abstract
Objective.The photo injector test facility at DESY in Zeuthen (PITZ) is building up an R&D platform, known as FLASHlab@PITZ, for systematically studying the FLASH effect in cancer treatment with its high-brightness electron beams, which can provide a uniquely large dose parameter range for radiation experiments. In this paper, we demonstrate the capabilities by experiments with a reduced parameter range on a startup beamline and study the potential performance of the full beamline by simulations.Approach.To measure the dose, Gafchromic films are installed both in front of and after the samples; Monte Carlo simulations are conducted to predict the dose distribution during beam preparation and help understand the dose distribution inside the sample. Plasmid DNA is irradiated under various doses at conventional and ultra-high dose rate (UHDR) to study the DNA damage by radiations. Start-to-end simulations are performed to verify the performance of the full beamline.Main results.On the startup beamline, reproducible irradiation has been established with optimized electron beams and the delivered dose distributions have been measured with Gafchromic films and compared to FLUKA simulations. The functionality of this setup has been further demonstrated in biochemical experiments at conventional dose rate of 0.05 Gy s-1and UHDR of several 105 Gy s-1and a varying dose up to 60 Gy, with the UHDR experiments finished within a single RF pulse (less than 1 millisecond); the observed conformation yields of the irradiated plasmid DNA revealed its dose-dependent radiation damage. The upgrade to the full FLASHlab@PITZ beamline is justified by simulations with homogeneous radiation fields generated by both pencil beam scanning and scattering beams.Significance.With the demonstration of UHDR irradiation and the simulated performance of the new beamline, FLASHlab@PITZ will serve as a powerful platform for studying the FLASH effects in cancer treatment.
Collapse
Affiliation(s)
- X-K Li
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - Z Amirkhanyan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - A Grebinyk
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - M Gross
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - Y Komar
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - F Riemer
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - A Asoyan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - P Boonpornprasert
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - P Borchert
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - H Davtyan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - D Dmytriiev
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - M Frohme
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - A Hoffmann
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - M Krasilnikov
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - G Loisch
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Z Lotfi
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - F Müller
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - M Schmitz
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - F Obier
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - A Oppelt
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - S Philipp
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - C Richard
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - G Vashchenko
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - D Villani
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - S Worm
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - F Stephan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| |
Collapse
|
5
|
P J N, Patil SR, Veeraraghavan VP, Daniel S, Aileni KR, Karobari MI. Oral cancer stem cells: A comprehensive review of key drivers of treatment resistance and tumor recurrence. Eur J Pharmacol 2025; 989:177222. [PMID: 39755243 DOI: 10.1016/j.ejphar.2024.177222] [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: 10/05/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025]
Abstract
Oral squamous cell carcinoma (OSCC) remains a major cause of morbidity and mortality worldwide with high recurrence rates and resistance to conventional therapies. Recent studies have highlighted the pivotal role of oral cancer stem cells (OCSCs) in driving treatment resistance and tumor recurrence. OCSCs possess unique properties, including self-renewal, differentiation potential, and resistance to chemotherapy and radiotherapy, which contribute to their ability to survive treatment and initiate tumor relapse. Several signaling pathways, such as Wnt/β-catenin, Hedgehog, Notch, and PI3K/Akt/mTOR, have been implicated in maintaining OCSC properties, promoting survival, and conferring resistance. Additionally, mechanisms such as drug efflux, enhanced DNA repair, epithelial-mesenchymal transition (EMT), and resistance to apoptosis further contribute to resilience. Targeting these pathways offers promising therapeutic strategies for eliminating OCSCs and improving treatment outcomes. Approaches such as immunotherapy, nanotechnology-based drug delivery, and targeting of the tumor microenvironment are emerging as potential solutions to overcome OCSC-mediated resistance. However, further research is needed to fully understand the molecular mechanisms governing OCSCs and develop effective therapies to prevent tumor recurrence. This review discusses the role of OCSCs in treatment resistance and recurrence and highlights the current and future directions for targeting these cells in OSCC.
Collapse
Affiliation(s)
- Nagarathna P J
- Department of Pediatric Dentistry, Chhattisgarh Dental College and Research Institute, India.
| | - Santosh R Patil
- Department of Oral Medicine and Radiology, Chhattisgarh Dental College and Research Institute, Rajnandgaon, C.G, India.
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India.
| | - Shikhar Daniel
- Department of Oral Medicine and Radiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India.
| | - Kaladhar Reddy Aileni
- Department of Preventive Dentistry, College of Dentistry, Jouf University, Chennai, Tamil Nadu, India.
| | - Mohmed Isaqali Karobari
- Department of Conservative Dentistry & Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
6
|
Kilmister EJ, Tan ST. Cancer Stem Cells and the Renin-Angiotensin System in the Tumor Microenvironment of Melanoma: Implications on Current Therapies. Int J Mol Sci 2025; 26:1389. [PMID: 39941158 PMCID: PMC11818896 DOI: 10.3390/ijms26031389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Multiple signaling pathways are dysregulated in melanoma, notably the Ras/RAF/MAPK/ERK and PI3K/AKT/mTOR pathways, which can be targeted therapeutically. The high immunogenicity of melanoma has been exploited using checkpoint inhibitors. Whilst targeted therapies and immune checkpoint inhibitors have improved the survival of patients with advanced melanoma, treatment resistance, their side effect profiles, and the prohibitive cost remain a challenge, and the survival outcomes remain suboptimal. Treatment resistance has been attributed to the presence of cancer stem cells (CSCs), a small subpopulation of pluripotent, highly tumorigenic cells proposed to drive cancer progression, recurrence, metastasis, and treatment resistance. CSCs reside within the tumor microenvironment (TME) regulated by the immune system, and the paracrine renin-angiotensin system, which is expressed in many cancer types, including melanoma. This narrative review discusses the role of CSCs and the paracrine renin-angiotensin system in the melanoma TME, and its implications on the current treatment of advanced melanoma with targeted therapy and immune checkpoint blockers. It also highlights the regulation of the Ras/RAF/MAPK/ERK and PI3K/AKT/mTOR pathways by the renin-angiotensin system via pro-renin receptors, and how this may relate to CSCs and treatment resistance, underscoring the potential for improving the efficacy of targeted therapy and immunotherapy by concurrently modulating the renin-angiotensin system.
Collapse
Affiliation(s)
- Ethan J. Kilmister
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand
- Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Lower Hutt 5010, New Zealand
| | - Swee T. Tan
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand
- Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Lower Hutt 5010, New Zealand
- Department of Surgery, The University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| |
Collapse
|
7
|
Li A, Wang R. Inhibition of Src Attenuates Stemness and Reverses Cisplatin Resistance of Non-Small-Cell Lung Cancer Cells. Bull Exp Biol Med 2025; 178:460-466. [PMID: 40156745 DOI: 10.1007/s10517-025-06356-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Indexed: 04/01/2025]
Abstract
The first-line chemical drug cisplatin used to treat the non-small cell lung cancer (NSCLC) often becomes ineffective due to acquired drug resistance of cancer cells. This effect and related cancer progression are related to the presence of cancer stem cells in the tumor. Tyrosine kinase Src is responsible for the appearance and development of cancer stem cells. Here, we examined the effects of Src on the oncogenic properties of cisplatin-resistant NSCLC cell lines (H358R and A549R) and the effect of inhibition of this tyrosine kinase on the sensitivity of cancer cells to cisplatin by Western blotting and immunofluorescence staining as well as migration, invasion, sphere colony, and clone formation assays. In H358R and A549R cells, the levels of phosphorylated kinase Src (pSrc) and stemness marker CD133 were elevated in comparison with the parental ones. The cisplatin-resistant cells demonstrated increased self-renewal ability and formed significantly bigger tumor spheres than their parental cells. Inhibition of Src with its inhibitor CGP77675 attenuated stemness of H358R and A549R cells; moreover, it enhanced the inhibitory effects of cisplatin on cell proliferation, migration, and invasion. The data indicated that Src-induced stemness plays an important role in developed resistance of NSCLC cells to cisplatin. Inhibition of Src attenuated stemness and acquired resistance to cisplatin, which can be beneficial for treating the cisplatin-resistant NSCLC.
Collapse
Affiliation(s)
- A Li
- Medical School, Anhui University of Science & Technology, Huainan, China
| | - R Wang
- Pulmonary and Critical Care Medicine, First Affiliated Hospital, Anhui University of Science & Technology (Huainan First People's Hospital), Huainan, China.
| |
Collapse
|
8
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
9
|
Molnár AÁ, Birgés K, Surman A, Merkely B. The Complex Connection Between Myocardial Dysfunction and Cancer Beyond Cardiotoxicity: Shared Risk Factors and Common Molecular Pathways. Int J Mol Sci 2024; 25:13185. [PMID: 39684895 DOI: 10.3390/ijms252313185] [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: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiovascular diseases and cancer represent the largest disease burden worldwide. Previously, these two conditions were considered independent, except in terms of cardiotoxicity, which links cancer treatment to subsequent cardiovascular issues. However, recent studies suggest that there are further connections between cancer and heart disease beyond cardiotoxicity. It has been revealed that myocardial dysfunction may promote carcinogenesis, indicating that additional common pathophysiological mechanisms might be involved in the relationship between cardiology and oncology, rather than simply a connection through cardiotoxic effects. These mechanisms may include shared risk factors and common molecular pathways, such as persistent inflammation and neurohormonal activation. This review explores the connection between myocardial dysfunction and cancer, emphasizing their shared risk factors, similar biological mechanisms, and causative factors like cardiotoxicity, along with their clinical implications.
Collapse
Affiliation(s)
| | - Kristóf Birgés
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Adrienn Surman
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| |
Collapse
|
10
|
Saranya I, Preetha D, Nivruthi S, Selvamurugan N. A comprehensive bioinformatic analysis of the role of TGF-β1-stimulated activating transcription factor 3 by non-coding RNAs during breast cancer progression. Comput Biol Chem 2024; 113:108208. [PMID: 39276678 DOI: 10.1016/j.compbiolchem.2024.108208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
A potent growth inhibitor for normal mammary epithelial cells is transforming growth factor beta 1 (TGF-β1). When breast tissues lose the anti-proliferative activity of this factor, invasion and bone metastases increase. Human breast cancer (hBC) cells express more activating transcription factor 3 (ATF3) when exposed to TGF-β1, and this transcription factor is essential for BC development and bone metastases. Non-coding RNAs (ncRNAs), including circular RNAs (circRNAs) and microRNAs (miRNAs), have emerged as key regulators controlling several cellular processes. In hBC cells, TGF-β1 stimulated the expression of hsa-miR-4653-5p that putatively targets ATF3. Bioinformatics analysis predicted that hsa-miR-4653-5p targets several key signaling components and transcription factors, including NFKB1, STAT1, STAT3, NOTCH1, JUN, TCF3, p300, NRF2, SUMO2, and NANOG, suggesting the diversified role of hsa-miR-4653-5p under physiological and pathological conditions. Despite the high abundance of hsa-miR-4653-5p in hBC cells, the ATF3 level remained elevated, indicating other ncRNAs could inhibit hsa-miR-4653-5p's activity. In silico analysis identified several circRNAs having the binding sites for hsa-miR-4653-5p, indicating the sponging activity of circRNAs towards hsa-miR-4653-5p. The study's findings suggest that TGF-β1 regulates circRNAs and hsa-miR-4653-5p, which in turn affects ATF3 expression, thus influencing BC progression and bone metastasis. Therefore, focusing on the TGF-β1/circRNAs/hsa-miR-4653-5p/ATF3 network could lead to new ways of diagnosing and treating BC.
Collapse
Affiliation(s)
- Iyyappan Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Dilipkumar Preetha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Sasi Nivruthi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India.
| |
Collapse
|
11
|
Rodrigues P, Bangali H, Ali E, Sharma MK, Abdullaev B, Alkhafaji AT, Deorari MM, Zabibah RS, Haslany A. Microproteins/micropeptides dysregulation contributes to cancer progression and development: A mechanistic review. Cell Biol Int 2024; 48:1395-1405. [PMID: 39010637 DOI: 10.1002/cbin.12219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 05/06/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
Microproteins, known as micropeptides, are small protein molecules encoded by short open reading frames. These recently identified molecules have been proven to be an essential part of the human proteome that participates in multiple processes, such as DNA repair, mitochondrial respiration, and regulating different signaling pathways. A growing body of studies has evidenced that microproteins exhibit dysregulated expression levels in various malignancies and contribute to tumor progression. It has been reported that microproteins interact with many proteins, such as enzymes (e.g., adenosine triphosphate synthase) and signal transducers (e.g., c-Jun), and regulate malignant cell metabolism, proliferation, and metastasis. Moreover, microproteins have been found to play a significant role in multidrug resistance in vitro and in vivo by their activity in DNA repair pathways. Considering that, this review intended to summarize the roles of microproteins in different aspects of tumorigenesis with diagnostic and therapeutic perspectives.
Collapse
Affiliation(s)
- Paul Rodrigues
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Harun Bangali
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Saudi Arabia
| | - Eyhab Ali
- College of Chemistry, Al-Zahraa University for Women, Karbala, Iraq
| | - M K Sharma
- Chaudhary Charan Singh University, Meerut, Uttar Pradesh, India
| | - Bekhzod Abdullaev
- Department of Biotechnology, New Uzbekistan University, Tashkent, Uzbekistan
| | | | - Maha Medha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Rahman S Zabibah
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Haslany
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| |
Collapse
|
12
|
Buccioli G, Testa C, Jacchetti E, Pinoli P, Carelli S, Ceri S, Raimondi MT. The molecular basis of the anticancer effect of statins. Sci Rep 2024; 14:20298. [PMID: 39217242 PMCID: PMC11365972 DOI: 10.1038/s41598-024-71240-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Statins, widely used cardiovascular drugs that lower cholesterol by inhibiting HMG-CoA reductase, have been increasingly recognized for their potential anticancer properties. This study elucidates the underlying mechanism, revealing that statins exploit Synthetic Lethality, a principle where the co-occurrence of two non-lethal events leads to cell death. Our computational analysis of approximately 37,000 SL pairs identified statins as potential drugs targeting genes involved in SL pairs with metastatic genes. In vitro validation on various cancer cell lines confirmed the anticancer efficacy of statins. This data-driven drug repurposing strategy provides a molecular basis for the anticancer effects of statins, offering translational opportunities in oncology.
Collapse
Affiliation(s)
- Giovanni Buccioli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Carolina Testa
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Pietro Pinoli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milan, Italy
| | - Stefano Ceri
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy.
| | - Manuela T Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy.
| |
Collapse
|
13
|
Brignola C, Pecoraro A, Danisi C, Iaccarino N, Di Porzio A, Romano F, Carotenuto P, Russo G, Russo A. uL3 Regulates Redox Metabolism and Ferroptosis Sensitivity of p53-Deleted Colorectal Cancer Cells. Antioxidants (Basel) 2024; 13:757. [PMID: 39061826 PMCID: PMC11274089 DOI: 10.3390/antiox13070757] [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: 06/03/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Despite advancements in therapeutic strategies, the development of drug resistance and metastasis remains a serious concern for the efficacy of chemotherapy against colorectal cancer (CRC). We have previously demonstrated that low expression of ribosomal protein uL3 positively correlates with chemoresistance in CRC patients. Here, we demonstrated that the loss of uL3 increased the metastatic capacity of CRC cells in chick embryos. Metabolomic analysis revealed large perturbations in amino acid and glutathione metabolism in resistant uL3-silenced CRC cells, indicating that uL3 silencing dramatically triggered redox metabolic reprogramming. RNA-Seq data revealed a notable dysregulation of 108 genes related to ferroptosis in CRC patients. Solute Carrier Family 7 Member 11 (SLC7A11) is one of the most dysregulated genes; its mRNA stability is negatively regulated by uL3, and its expression is inversely correlated with uL3 levels. Inhibition of SLC7A11 with erastin impaired resistant uL3-silenced CRC cell survival by inducing ferroptosis. Of interest, the combined treatment erastin plus uL3 enhanced the chemotherapeutic sensitivity of uL3-silenced CRC cells to erastin. The antimetastatic potential of the combined strategy was evaluated in chick embryos. Overall, our study sheds light on uL3-mediated chemoresistance and provides evidence of a novel therapeutic approach, erastin plus uL3, to induce ferroptosis, establishing individualized therapy by examining p53, uL3 and SLC7A11 profiles in tumors.
Collapse
Affiliation(s)
- Chiara Brignola
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| | - Annalisa Pecoraro
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| | - Camilla Danisi
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| | - Nunzia Iaccarino
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| | - Anna Di Porzio
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| | - Francesca Romano
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| | - Pietro Carotenuto
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei, 34, 80078 Naples, Italy;
- Medical Genetics, Department of Translational Medical Science, University of Naples “Federico II”, Corso Umberto I, 40, 80138 Naples, Italy
| | - Giulia Russo
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| | - Annapina Russo
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.B.); (A.P.); (C.D.); (N.I.); (A.D.P.); (F.R.); (G.R.)
| |
Collapse
|
14
|
Mazzoccoli L, Liu B. Dendritic Cells in Shaping Anti-Tumor T Cell Response. Cancers (Basel) 2024; 16:2211. [PMID: 38927916 PMCID: PMC11201542 DOI: 10.3390/cancers16122211] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Among professional antigen-presenting cells, dendritic cells (DCs) orchestrate innate and adaptive immunity and play a pivotal role in anti-tumor immunity. DCs are a heterogeneous population with varying functions in the tumor microenvironment (TME). Tumor-associated DCs differentiate developmentally and functionally into three main subsets: conventional DCs (cDCs), plasmacytoid DCs (pDCs), and monocyte-derived DCs (MoDCs). There are two major subsets of cDCs in TME, cDC1 and cDC2. cDC1 is critical for cross-presenting tumor antigens to activate cytotoxic CD8+ T cells and is also required for priming earlier CD4+ T cells in certain solid tumors. cDC2 is vital for priming anti-tumor CD4+ T cells in multiple tumor models. pDC is a unique subset of DCs and produces type I IFN through TLR7 and TLR9. Studies have shown that pDCs are related to immunosuppression in the TME through the secretion of immunosuppressive cytokines and by promoting regulatory T cells. MoDCs differentiate separately from monocytes in response to inflammatory cues and infection. Also, MoDCs can cross-prime CD8+ T cells. In this review, we summarize the subsets and functions of DCs. We also discuss the role of different DC subsets in shaping T cell immunity in TME and targeting DCs for potential immunotherapeutic benefits against cancer.
Collapse
Affiliation(s)
- Luciano Mazzoccoli
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Siddique R, Thangavelu L, S R, Almalki WH, Kazmi I, Kumar A, Mahajan S, Kalra H, Alzarea SI, Pant K. lncRNAs and cyclin-dependent kinases: Unveiling their critical roles in cancer progression. Pathol Res Pract 2024; 258:155333. [PMID: 38723325 DOI: 10.1016/j.prp.2024.155333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024]
Abstract
Long non-coding RNAs (lncRNAs) are a diverse class of RNA molecules that do not code for proteins but play critical roles in gene regulation. One such role involves the modulation of cell cycle progression and proliferation through interactions with cyclin-dependent kinases (CDKs), key regulators of cell division. Dysregulation of CDK activity is a hallmark of cancer, contributing to uncontrolled cell growth and tumor formation. These lncRNA-CDK interactions are part of a complex network of molecular mechanisms underlying cancer pathogenesis, involving various signaling pathways and regulatory circuits. Understanding the interplay between lncRNAs, CDKs, and cancer biology holds promise for developing novel therapeutic strategies targeting these molecular targets for more effective cancer treatment. Furthermore, targeting CDKs, key cell cycle progression and proliferation regulators, offers another avenue for disrupting cancer pathways and overcoming drug resistance. This can open new possibilities for individualized treatment plans and focused therapeutic interventions.
Collapse
Affiliation(s)
- Raihan Siddique
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Lakshmi Thangavelu
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India.
| | - RenukaJyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Ashwani Kumar
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Shriya Mahajan
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140417, India
| | - Hitesh Kalra
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh 174103, India
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Al-Jouf, Saudi Arabia
| | - Kumud Pant
- Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India; Graphic Era Hill University, Clement Town, Dehradun 248002, India
| |
Collapse
|
16
|
Cao J, Li C, Cui Z, Deng S, Lei T, Liu W, Yang H, Chen P. Spatial Transcriptomics: A Powerful Tool in Disease Understanding and Drug Discovery. Theranostics 2024; 14:2946-2968. [PMID: 38773973 PMCID: PMC11103497 DOI: 10.7150/thno.95908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/25/2024] [Indexed: 05/24/2024] Open
Abstract
Recent advancements in modern science have provided robust tools for drug discovery. The rapid development of transcriptome sequencing technologies has given rise to single-cell transcriptomics and single-nucleus transcriptomics, increasing the accuracy of sequencing and accelerating the drug discovery process. With the evolution of single-cell transcriptomics, spatial transcriptomics (ST) technology has emerged as a derivative approach. Spatial transcriptomics has emerged as a hot topic in the field of omics research in recent years; it not only provides information on gene expression levels but also offers spatial information on gene expression. This technology has shown tremendous potential in research on disease understanding and drug discovery. In this article, we introduce the analytical strategies of spatial transcriptomics and review its applications in novel target discovery and drug mechanism unravelling. Moreover, we discuss the current challenges and issues in this research field that need to be addressed. In conclusion, spatial transcriptomics offers a new perspective for drug discovery.
Collapse
Affiliation(s)
- Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| | - Tong Lei
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wei Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| |
Collapse
|
17
|
Chung YH, Zhao Z, Jung E, Omole AO, Wang H, Sutorus L, Steinmetz NF. Systemic Administration of Cowpea Mosaic Virus Demonstrates Broad Protection Against Metastatic Cancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308237. [PMID: 38430536 PMCID: PMC11095214 DOI: 10.1002/advs.202308237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/21/2023] [Indexed: 03/04/2024]
Abstract
The key challenge in cancer treatment is prevention of metastatic disease which is therapeutically resistant and carries poor prognoses necessitating efficacious prophylactic approaches that prevent metastasis and recurrence. It is previously demonstrated that cowpea mosaic virus (CPMV) induces durable antitumor responses when used in situ, i.e., intratumoral injection. As a new direction, it is showed that CPMV demonstrates widespread effectiveness as an immunoprophylactic agent - potent efficacy is demonstrated in four metastatic models of colon, ovarian, melanoma, and breast cancer. Systemic administration of CPMV stimulates the innate immune system, enabling attack of cancer cells; processing of the cancer cells and associated antigens leads to systemic, durable, and adaptive antitumor immunity. Overall, CPMV demonstrated broad efficacy as an immunoprophylactic agent in the rejection of metastatic cancer.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Moores Cancer CenterUniversity of California, San DiegoLa JollaCA92093USA
| | - Zhongchao Zhao
- Moores Cancer CenterUniversity of California, San DiegoLa JollaCA92093USA
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
- Center for Nano‐ImmunoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Eunkyeong Jung
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Anthony O. Omole
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Hanyang Wang
- Department of BiologyUniversity of California, San DiegoLa JollaCA92093USA
| | - Lucas Sutorus
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
| | - Nicole F. Steinmetz
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Moores Cancer CenterUniversity of California, San DiegoLa JollaCA92093USA
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
- Center for Nano‐ImmunoEngineeringUniversity of California, San DiegoLa JollaCA92093USA
- Department of RadiologyUniversity of California, San DiegoLa JollaCA92093USA
- Institute for Materials Discovery and DesignUniversity of California, San DiegoLa JollaCA92093USA
- Center for Engineering in CancerUniversity of California, San DiegoLa JollaCA92093USA
- Shu and K.C. Chien and Peter Farrell CollaboratoryUniversity of California, San DiegoLa JollaCA92093USA
| |
Collapse
|
18
|
Weth FR, Hoggarth GB, Weth AF, Paterson E, White MPJ, Tan ST, Peng L, Gray C. Unlocking hidden potential: advancements, approaches, and obstacles in repurposing drugs for cancer therapy. Br J Cancer 2024; 130:703-715. [PMID: 38012383 PMCID: PMC10912636 DOI: 10.1038/s41416-023-02502-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023] Open
Abstract
High rates of failure, exorbitant costs, and the sluggish pace of new drug discovery and development have led to a growing interest in repurposing "old" drugs to treat both common and rare diseases, particularly cancer. Cancer, a complex and heterogeneous disease, often necessitates a combination of different treatment modalities to achieve optimal outcomes. The intrinsic polygenicity of cancer, intricate biological signalling networks, and feedback loops make the inhibition of a single target frequently insufficient for achieving the desired therapeutic impact. As a result, addressing these complex or "smart" malignancies demands equally sophisticated treatment strategies. Combinatory treatments that target the multifaceted oncogenic signalling network hold immense promise. Repurposed drugs offer a potential solution to this challenge, harnessing known compounds for new indications. By avoiding the prohibitive costs and long development timelines associated with novel cancer drugs, this approach holds the potential to usher in more effective, efficient, and cost-effective cancer treatments. The pursuit of combinatory therapies through drug repurposing may hold the key to achieving superior outcomes for cancer patients. However, drug repurposing faces significant commercial, technological and regulatory challenges that need to be addressed. This review explores the diverse approaches employed in drug repurposing, delves into the challenges faced by the drug repurposing community, and presents innovative solutions to overcome these obstacles. By emphasising the significance of combinatory treatments within the context of drug repurposing, we aim to unlock the full potential of this approach for enhancing cancer therapy. The positive aspects of drug repurposing in oncology are underscored here; encompassing personalized treatment, accelerated development, market opportunities for shelved drugs, cancer prevention, expanded patient reach, improved patient access, multi-partner collaborations, increased likelihood of approval, reduced costs, and enhanced combination therapy.
Collapse
Affiliation(s)
- Freya R Weth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | - Georgia B Hoggarth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | - Anya F Weth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | - Erin Paterson
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | | | - Swee T Tan
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt, 5040, New Zealand
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
19
|
Kast RE. The OSR9 Regimen: A New Augmentation Strategy for Osteosarcoma Treatment Using Nine Older Drugs from General Medicine to Inhibit Growth Drive. Int J Mol Sci 2023; 24:15474. [PMID: 37895152 PMCID: PMC10607234 DOI: 10.3390/ijms242015474] [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: 09/23/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
As things stand in 2023, metastatic osteosarcoma commonly results in death. There has been little treatment progress in recent decades. To redress the poor prognosis of metastatic osteosarcoma, the present regimen, OSR9, uses nine already marketed drugs as adjuncts to current treatments. The nine drugs in OSR9 are: (1) the antinausea drug aprepitant, (2) the analgesic drug celecoxib, (3) the anti-malaria drug chloroquine, (4) the antibiotic dapsone, (5) the alcoholism treatment drug disulfiram, (6) the antifungal drug itraconazole, (7) the diabetes treatment drug linagliptin, (8) the hypertension drug propranolol, and (9) the psychiatric drug quetiapine. Although none are traditionally used to treat cancer, all nine have attributes that have been shown to inhibit growth-promoting physiological systems active in osteosarcoma. In their general medicinal uses, all nine drugs in OSR9 have low side-effect risks. The current paper reviews the collected data supporting the role of OSR9.
Collapse
|
20
|
Ashekyan O, Shahbazyan N, Bareghamyan Y, Kudryavzeva A, Mandel D, Schmidt M, Loeffler-Wirth H, Uduman M, Chand D, Underwood D, Armen G, Arakelyan A, Nersisyan L, Binder H. Transcriptomic Maps of Colorectal Liver Metastasis: Machine Learning of Gene Activation Patterns and Epigenetic Trajectories in Support of Precision Medicine. Cancers (Basel) 2023; 15:3835. [PMID: 37568651 PMCID: PMC10417131 DOI: 10.3390/cancers15153835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The molecular mechanisms of the liver metastasis of colorectal cancer (CRLM) remain poorly understood. Here, we applied machine learning and bioinformatics trajectory inference to analyze a gene expression dataset of CRLM. We studied the co-regulation patterns at the gene level, the potential paths of tumor development, their functional context, and their prognostic relevance. Our analysis confirmed the subtyping of five liver metastasis subtypes (LMS). We provide gene-marker signatures for each LMS, and a comprehensive functional characterization that considers both the hallmarks of cancer and the tumor microenvironment. The ordering of CRLMs along a pseudotime-tree revealed a continuous shift in expression programs, suggesting a developmental relationship between the subtypes. Notably, trajectory inference and personalized analysis discovered a range of epigenetic states that shape and guide metastasis progression. By constructing prognostic maps that divided the expression landscape into regions associated with favorable and unfavorable prognoses, we derived a prognostic expression score. This was associated with critical processes such as epithelial-mesenchymal transition, treatment resistance, and immune evasion. These factors were associated with responses to neoadjuvant treatment and the formation of an immuno-suppressive, mesenchymal state. Our machine learning-based molecular profiling provides an in-depth characterization of CRLM heterogeneity with possible implications for treatment and personalized diagnostics.
Collapse
Affiliation(s)
- Ohanes Ashekyan
- Armenian Bioinformatics Institute, 3/6 Nelson Stepanyan Str., Yerevan 0062, Armenia; (O.A.); (N.S.); (Y.B.); (A.K.); (D.M.); (L.N.)
| | - Nerses Shahbazyan
- Armenian Bioinformatics Institute, 3/6 Nelson Stepanyan Str., Yerevan 0062, Armenia; (O.A.); (N.S.); (Y.B.); (A.K.); (D.M.); (L.N.)
| | - Yeva Bareghamyan
- Armenian Bioinformatics Institute, 3/6 Nelson Stepanyan Str., Yerevan 0062, Armenia; (O.A.); (N.S.); (Y.B.); (A.K.); (D.M.); (L.N.)
| | - Anna Kudryavzeva
- Armenian Bioinformatics Institute, 3/6 Nelson Stepanyan Str., Yerevan 0062, Armenia; (O.A.); (N.S.); (Y.B.); (A.K.); (D.M.); (L.N.)
| | - Daria Mandel
- Armenian Bioinformatics Institute, 3/6 Nelson Stepanyan Str., Yerevan 0062, Armenia; (O.A.); (N.S.); (Y.B.); (A.K.); (D.M.); (L.N.)
| | - Maria Schmidt
- IZBI, Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany; (M.S.); (H.L.-W.)
| | - Henry Loeffler-Wirth
- IZBI, Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany; (M.S.); (H.L.-W.)
| | - Mohamed Uduman
- Agenus Inc., 3 Forbes Road, Lexington, MA 7305, USA; (M.U.); (D.C.); (D.U.); (G.A.)
| | - Dhan Chand
- Agenus Inc., 3 Forbes Road, Lexington, MA 7305, USA; (M.U.); (D.C.); (D.U.); (G.A.)
| | - Dennis Underwood
- Agenus Inc., 3 Forbes Road, Lexington, MA 7305, USA; (M.U.); (D.C.); (D.U.); (G.A.)
| | - Garo Armen
- Agenus Inc., 3 Forbes Road, Lexington, MA 7305, USA; (M.U.); (D.C.); (D.U.); (G.A.)
| | - Arsen Arakelyan
- Institute of Molecular Biology of the National Academy of Sciences of the Republic of Armenia, 7 Has-Ratyan Str., Yerevan 0014, Armenia;
| | - Lilit Nersisyan
- Armenian Bioinformatics Institute, 3/6 Nelson Stepanyan Str., Yerevan 0062, Armenia; (O.A.); (N.S.); (Y.B.); (A.K.); (D.M.); (L.N.)
| | - Hans Binder
- Armenian Bioinformatics Institute, 3/6 Nelson Stepanyan Str., Yerevan 0062, Armenia; (O.A.); (N.S.); (Y.B.); (A.K.); (D.M.); (L.N.)
- IZBI, Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany; (M.S.); (H.L.-W.)
| |
Collapse
|
21
|
Hairunisa I, Bakar MFA, Da'i M, Bakar FIA, Syamsul ES. Cytotoxic Activity, Anti-Migration and In Silico Study of Black Ginger ( Kaempferia parviflora) Extract against Breast Cancer Cell. Cancers (Basel) 2023; 15:2785. [PMID: 37345122 DOI: 10.3390/cancers15102785] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 06/23/2023] Open
Abstract
Metastatic breast cancer remains the leading cause of death in women worldwide. This condition necessitates extensive research to find an effective treatment, one of which is the natural medicine approach. Kaempferia parviflora (KP) is a plant believed to possess anticancer properties. Therefore, this study aims to determine KP's bioactive compound, cytotoxic, and anti-migration activity in the highly metastatic breast cancer cell line model 4T1, also in the breast cancer cell model MCF-7 and noncancerous cell line NIH-3T3. Maceration with ethanol (EEKP) and infusion with distilled water (EWKP) was used for extraction. The MTT assay was used to test for cytotoxicity, and the scratch wound healing assay was used to test for the inhibition of migration. Phytochemical profiling of EEKP was performed using UHPLC-MS, and the results were studied for in silico molecular docking. Result showed that EEKP had a better cytotoxic activity than EWKP with an IC50 value of 128.33 µg/mL (24 h) and 115.09 µg/mL (48 h) on 4T1 cell line, and 138.43 µg/mL (24 h) and 124.81 µg/mL (48 h) on MCF-7 cell line. Meanwhile, no cytotoxic activity was observed at concentrations ranging from 3-250 µg/mL in NIH-3T3. EEKP also showed anti-migration activity in a concentration of 65 µg/mL. Mass Spectrophotometer (MS) structures from EEKP are 5-Hydroxy-7,4'-dimethoxyflavanone (HDMF), 5-Hydro-7,8,2'-trimethoxyflavanone (HTMF), Retusine, and Denbinobin. The in silico docking was investigated for receptors Bcl-2, Bcl-XL, ERK2, and FAK, as well as their activities. In silico result indicates that HTMF and denbinobin are bioactive compounds responsible for EEKP's cytotoxic and anti-migration activity. These two compounds and standardized plant extract can be further studied as potential breast cancer treatment candidates.
Collapse
Affiliation(s)
- Indah Hairunisa
- Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia (UTHM), Muar 84600, Malaysia
- Faculty of Pharmacy, Universitas Muhammadiyah Kalimantan Timur (UMKT), Samarinda 75124, Indonesia
| | - Mohd Fadzelly Abu Bakar
- Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia (UTHM), Muar 84600, Malaysia
| | - Muhammad Da'i
- Faculty of Pharmacy, Universitas Muhammadiyah Surakarta (UMS), Solo 57162, Indonesia
| | - Fazleen Izzany Abu Bakar
- Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia (UTHM), Muar 84600, Malaysia
| | | |
Collapse
|
22
|
Yeh ES. Special Issue: Cancer Metastasis and Therapeutic Resistance. Biomedicines 2023; 11:biomedicines11051347. [PMID: 37239018 DOI: 10.3390/biomedicines11051347] [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: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Metastasis and resistance to cancer therapeutics are critical barriers to curing cancer. This special issue entitled "Cancer Metastasis and Therapeutic Resistance" contains nine original contributions. The articles span a variety of human cancers, including breast, lung, brain, prostate, and skin and touch upon significant areas of interest such as cancer stem cell function, cancer immunology, and glycosylation.
Collapse
Affiliation(s)
- Elizabeth S Yeh
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|