1
|
Bollu VS, Chen YC, Zhang F, Gowda K, Amin S, Sharma AK, Schell TD, Zhu J, Robertson GP. Managing telomerase and telomere dysfunction in acral melanoma. Pharmacol Res 2025; 215:107700. [PMID: 40097124 DOI: 10.1016/j.phrs.2025.107700] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Acral Lentiginous Melanoma is a rare and aggressive subtype of melanoma that commonly affects the palms, soles, and nail beds. It is more prevalent in individuals with darker skin tones, including Asian, African, and Hispanic populations. Unlike cutaneous melanomas, acral melanoma is not associated with UV exposure and has a distinct genetic and molecular profile, underscoring the need for tailored research and treatment strategies. Standard treatments, such as surgery, chemotherapy, immunotherapy, and targeted therapies, have shown limited success for this melanoma subtype, highlighting the urgency of developing more effective interventions. Telomerase is an enzyme that extends telomeres and is a key target in acral melanoma which exhibits' high telomerase activity, driven by mutations in the telomerase reverse transcriptase TERT promoter, which contributes to uncontrolled tumor cell proliferation, cancer cell immortality, and resistance to conventional therapies. Therefore, targeting telomerase presents a promising therapeutic avenue for acral melanoma patients who do not respond well to current treatments. Several approaches for targeting telomerase deregulation have been developed, and their potential for the management of acral melanoma is discussed in this review. Specifically, the promise of telomerase-targeted therapies for acral melanoma is emphasized and explores how these strategies could improve outcomes for patients with this challenging skin cancer. By focusing on the role of telomerase in tumorigenesis and treatment resistance, telomerase-targeted strategies hold potential as a foundational component of therapies for acral melanoma, complementing existing approaches.
Collapse
Affiliation(s)
- Vishnu Sravan Bollu
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Yu-Chi Chen
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Fan Zhang
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Krishne Gowda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Shantu Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Arun K Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Todd D Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Jiyue Zhu
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Gavin P Robertson
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Surgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma and Skin Cancer Center, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma Therapeutics Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
2
|
Kassie TD, Yimenu BW, Baye Temesgen G, Shimelash RA, Abneh AA. Differences in the count of blood cells pre-and post-chemotherapy in patients with cancer: a retrospective study (2022). Front Med (Lausanne) 2025; 12:1485676. [PMID: 40309726 PMCID: PMC12041017 DOI: 10.3389/fmed.2025.1485676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Cancer is a disease characterized by uncontrolled cell growth that can invade and spread to other body parts. Drugs used for chemotherapy can cause damage to non-cancer cells and lead to a low blood cell count. There are controversial findings regarding the differences in the counts of blood cell types. Studies on the counts of blood cell types before and after chemotherapy are limited. Therefore, this study aimed to address this gap. Methods A retrospective study was conducted on 354 patients from 1 September 2022 to 1 October 2022 to compare blood cell type profiles in pre-and post-operative periods. A chi-squared test and paired t-test were performed to analyze the data. Results Data were collected from 354 patients. The mean age of the respondents was 41.26 (±16.67) years. At the initial diagnosis, 167 patients (47%) were categorized as having stage III cancer, while 159 patients were categorized as having stage IV cancer. Before and after chemotherapy the mean heamoglobin level is 12.95 g/dL and 12.30 g/ dL respectively and the prevalence of anemia among cancer patients before chemotherapy and after chemotherapy was 25.14% (95%:20.88, 29.94) and 35.54% (95%: 30.75; 40.73), respectively. The mean value of neutrophils before and after chemotherapy was 52.72 and 50.82%, respectively. The frequency of neutropenia among cancer patients was 22.32% (95% 18.26; 26.96) before chemotherapy and 27.97% (95%:23.52; 32.88) after chemotherapy, and the mean value of lymphocytes before and after chemotherapy was 37.50 and 34.29%, respectively. Lymphopenia among the study participants was 16.38% (95%:12.87; 20.62) before chemotherapy and 17.51% (95%13.88, 21.84) after chemotherapy. Among patients with stomach, rectal, and bone cancers, there was no significant difference in the counts of all blood cell types before and after chemotherapy. The mean reduction in platelets was 23. 51 × 103cells/mm3 (p = 0.001). For red blood cells (RBCs), the mean decrease was 0.63 × 106cells/mm3 (p = 0.08), and for white blood cells (WBCs), it was 2.49 × 106 cells/mm3 (p = 0.012). Conclusion and recommendation After chemotherapy, all hematological parameters showed a decrease, indicating that chemotherapy significant impacted the levels of these hematological parameters. Therefore, replacement therapy should be considered after chemotherapy.
Collapse
Affiliation(s)
- Tadele Derbew Kassie
- Department of Public Health, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Bayu Wondimneh Yimenu
- Biomedical Department, College of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gelagey Baye Temesgen
- Biomedical Department, College of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Asres Shimelash
- Department of Nursing, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Aysheshim Asnake Abneh
- Department of Public Health, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
3
|
Gevertz JL, Greene JM, Prosperi S, Comandante-Lou N, Sontag ED. Understanding therapeutic tolerance through a mathematical model of drug-induced resistance. NPJ Syst Biol Appl 2025; 11:30. [PMID: 40204801 PMCID: PMC11982405 DOI: 10.1038/s41540-025-00511-3] [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: 09/03/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
There is growing recognition that phenotypic plasticity enables cancer cells to adapt to various environmental conditions. An example of this adaptability is the ability of an initially sensitive population of cancer cells to acquire resistance and persist in the presence of therapeutic agents. Understanding the implications of this drug-induced resistance is essential for predicting transient and long-term tumor dynamics subject to treatment. This paper introduces a mathematical model of drug-induced resistance which provides excellent fits to time-resolved in vitro experimental data. From observational data of total numbers of cells, the model unravels the relative proportions of sensitive and resistance subpopulations and quantifies their dynamics as a function of drug dose. The predictions are then validated using data on drug doses that were not used when fitting parameters. Optimal control techniques are then utilized to discover dosing strategies that could lead to better outcomes as quantified by lower total cell volume.
Collapse
Affiliation(s)
- Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA
| | - James M Greene
- Department of Mathematics, Clarkson University, Potsdam, NY, USA
| | - Samantha Prosperi
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Natacha Comandante-Lou
- Center for Translational & Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
| | - Eduardo D Sontag
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Kim HK, Cheong H, Kim MY, Jin HE. Therapeutic Targeting in Ovarian Cancer: Nano-Enhanced CRISPR/Cas9 Gene Editing and Drug Combination Therapy. Int J Nanomedicine 2025; 20:3907-3931. [PMID: 40191042 PMCID: PMC11970428 DOI: 10.2147/ijn.s507688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025] Open
Abstract
Ovarian cancer is the third most common gynecological cancer worldwide. Due to the high recurrence rate of advanced-stage ovarian cancer, often resulting from drug-resistant and refractory disease, various treatment strategies are under investigation. Genome editing of therapeutic target genes holds promise in enhancing cancer treatment efficacy by elucidating gene functions and mechanisms involved in cancer progression. The CRISPR/Cas9 system, in particular, shows great potential in ovarian cancer gene therapy and drug development. Targeting therapeutic genes such as BRCA1/2, P53, Snai1 etc, could improve the therapeutic strategy in ovarian cancer. CRISPR/Cas9 is a powerful gene-editing tool that there are many on-going clinical trials to treat various diseases including cancer. Nano-based delivery systems for CRISPR/Cas9 offer further therapeutic benefits, leveraging the unique properties of nanoparticles to improve delivery efficiency. Nano-based delivery systems could enhance the stability of CRISPR/Cas9 delivery formats (such as plasmid, mRNA, etc) and improve the delivery precision of delivery to target tumors. Additionally, combining CRISPR/Cas9 with targeted drug treatments, especially those aimed at genes associated with drug resistance, may significantly improve therapeutic outcomes in ovarian cancer. In this review, we discuss therapeutic target genes and their mechanisms in ovarian cancer, advances in nano-based CRISPR/Cas9 delivery, and the therapeutic potential of combining CRISPR/Cas9 with drug treatments for ovarian cancer.
Collapse
Affiliation(s)
- Hong-Kook Kim
- AI-Super Convergence KIURI Translational Research Center, Ajou University, Suwon, 16499, Republic of Korea
- Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Heedon Cheong
- Department of Pharmacy, Ajou University, Suwon, 16499, Republic of Korea
| | - Moo-Yeon Kim
- Department of Pharmacy, Ajou University, Suwon, 16499, Republic of Korea
| | - Hyo-Eon Jin
- Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
- Department of Pharmacy, Ajou University, Suwon, 16499, Republic of Korea
| |
Collapse
|
5
|
Arya BD, Mittal S, Joshi P, Pandey AK, Ramirez-Vick JE, Gupta G, Singh SP. Graphene oxide-chloroquine conjugate induces DNA damage in A549 lung cancer cells through autophagy modulation. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:316-332. [PMID: 40041432 PMCID: PMC11878127 DOI: 10.3762/bjnano.16.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025]
Abstract
Autophagy is a highly regulated catabolic process by which unnecessary, dysfunctional, or damaged proteins and other cellular components are degraded and recycled to promote cellular differentiation, survival, and development. In response to endogenous or exogenous stresses, cancer cells use autophagy pathways for survival through activation of complex DNA damage repair (DDR) mechanisms. In the present study, we demonstrated the genotoxicity induced in A549 lung cancer cells by exposure to the GO-Chl nanoconjugate and elucidated the role of autophagy modulation in harnessing the DNA-damage response. GO-Chl causes loss of plasma membrane integrity, cell cycle arrest, and significant genotoxicity in A549 cells. Further, elevated expression of key autophagy proteins beclin-1, ATG-7, LC-3-I/II, and SQSTM1/p62 reveal that inhibition of autophagy plays a crucial role in regulating DDR capabilities of cancer cells. The results indicate that the interplay between DDR and autophagy pathways may open new paradigms for developing effective combinatorial nanoscale drug systems against multidrug-resistance cancers.
Collapse
Affiliation(s)
- Braham Dutt Arya
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
- Department of Higher Education, Shiksha Sadan, Sector-5, Panchkula-134114, India
| | - Sandeep Mittal
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Mahatma Gandhi Marg, Lucknow-226001, India
| | - Prachi Joshi
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
| | - Alok Kumar Pandey
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Mahatma Gandhi Marg, Lucknow-226001, India
| | - Jaime E Ramirez-Vick
- Department of Biomedical, Industrial & Human Factors Engineering, Wright State University, Dayton, Ohio 45435, United States
| | - Govind Gupta
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
| | - Surinder P Singh
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
| |
Collapse
|
6
|
Gallenzi P, Malatesta A, Staderini E, Guglielmi F. Chemotherapy with Alkylating Agents and Dental Anomalies in Children: A Systematic Review. J Clin Med 2025; 14:1030. [PMID: 39941698 PMCID: PMC11818895 DOI: 10.3390/jcm14031030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
The aim of the systematic review is to analyze the type and the prevalence of dental side effects among cancer survivors treated with alkylating agents (AAs) during pediatric age. Moreover, the study aimed to investigate the association between the development of dental anomalies and the drug used or the tumor type. Four databases MEDLINE-PubMed, Web of Science, Scopus, and the Cochrane Central Register of Controlled Trials (CENTRAL) were searched from January 2024 to March 2024. All articles published up to March 2024 were evaluated. After removing duplicates, data extraction and risk of bias assessment using the Newcastle-Ottawa score were made. A summary of the overall strength of evidence available was performed using the "Grading of Recommendations Assessment, Development and Evaluation" (GRADE). Data were summarized using descriptive analysis as mean differences ± standard deviation or relative risks. Out of 2678 studies, the search identified five studies enrolled for the qualitative analysis of the data. Among 257 survivors, 155 (60.3%) reported: microdontia, agenesia, root shortening, enamel defects, and taurodontism. Microdontia occurred more frequently with other drugs compared to AAs. In conclusion, children treated with AAs showed microdontia (36.0%), root shortening (26.9%), and agenesis (23.5%). Secondly, the occurrence of dental anomalies was unaffected by drug treatment; thirdly, microdontia was the most frequent dental anomaly observed in both solid and lymphoproliferative tumors. This review was performed in accordance with the PRISMA guidelines. PROSPERO registration number CRD42023494560.
Collapse
Affiliation(s)
- Patrizia Gallenzi
- Institute of Dental Clinic, A. Gemelli University Policlinic IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (P.G.); (E.S.); (F.G.)
- Postgraduate School of Orthodontics, Director: Prof. Massimo Cordaro, Catholic University of Sacred Heart, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Angela Malatesta
- Institute of Dental Clinic, A. Gemelli University Policlinic IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (P.G.); (E.S.); (F.G.)
| | - Edoardo Staderini
- Institute of Dental Clinic, A. Gemelli University Policlinic IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (P.G.); (E.S.); (F.G.)
- Postgraduate School of Orthodontics, Director: Prof. Massimo Cordaro, Catholic University of Sacred Heart, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Federica Guglielmi
- Institute of Dental Clinic, A. Gemelli University Policlinic IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (P.G.); (E.S.); (F.G.)
- Postgraduate School of Orthodontics, Director: Prof. Massimo Cordaro, Catholic University of Sacred Heart, Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
7
|
Arvindh S, Priyadarshini M, Baba AB, Veeravarmal V, Mishra R, Dash R, Nagini S. The Neem Limonoid Nimbolide Modulates Key Components of the DNA Damage Response Signalling in Cellular and Animal Models of Oral Squamous Cell Carcinoma. Curr Pharm Biotechnol 2025; 26:428-442. [PMID: 38561609 DOI: 10.2174/0113892010291998240321074920] [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: 11/14/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Deregulated DNA damage response (DDR) network is implicated in cancer progression and therapy resistance. OBJECTIVE The present study was designed to investigate whether nimbolide, an anticancer neem limonoid, targets key components of the DDR signalling pathway in cellular and animal models of oral squamous cell carcinoma (OSCC). METHODS OSCC cells (SCC-4 and SCC-9), 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinoma model, chemoresistant OSCC patient-derived xenograft (PDX) model established in athymic nude mice, and tissue sections from patients with oral premalignant/malignant disease were used for the study. Key molecules that orchestrate the DDR, including the MRN complex, ATM, DNA-PKcs, H2AX, and p53, were analysed by qRTPCR, immunoblotting, immunofluorescence, and immunohistochemistry. Cell proliferation and apoptosis indices were evaluated. RESULTS Nimbolide significantly reduced 8-oxodG levels, expression of MRN, ATMS1891, and γ- H2AX, with an increase in p-p53S15 in OSCC cells as well as in the HBP model. Nimbolide potentiated the effect of KU-55933 in ATM inhibition. In the PDX model, nimbolide suppressed tumor formation, stimulated DDR and apoptosis, inhibited cell proliferation, and enhanced sensitivity to cisplatin. Analysis of p-ATM expression revealed a significant increase during the sequential progression of hamster and human OSCC. CONCLUSION This study provides compelling evidence that nimbolide functions as a DDR inhibitor in cellular and hamster OSCC models and as a DDR activator in the PDX model primarily by targeting ATM. Small molecules like nimbolide that modulate DDR are of immense benefit in cancer therapy. The study has also unveiled p-ATM as a promising biomarker of tumour progression in human OSCCs.
Collapse
Affiliation(s)
- Soundararajan Arvindh
- Department of Biochemistry & Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608002, Tamil Nadu, India
| | | | - Abdul Basit Baba
- Advanced Centre for Human Genetics, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu & Kashmir, 190011, India
| | - Veeran Veeravarmal
- Division of Oral and Maxillofacial Pathology, Government Dental College and Hospital, Cuddalore District, 608 002, Tamil Nadu, India
| | - Rajakishore Mishra
- Department of Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi, 835205, Jharkhand, India
| | - Rupesh Dash
- Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Siddavaram Nagini
- Department of Biochemistry & Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608002, Tamil Nadu, India
| |
Collapse
|
8
|
Elmorsy EA, Saber S, Hamad RS, Abdel-Reheim MA, El-Kott AF, AlShehri MA, Morsy K, Salama SA, Youssef ME. Advances in understanding cisplatin-induced toxicity: Molecular mechanisms and protective strategies. Eur J Pharm Sci 2024; 203:106939. [PMID: 39423903 DOI: 10.1016/j.ejps.2024.106939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/30/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Cisplatin, a widely used chemotherapeutic agent, has proven efficacy against various malignancies. However, its clinical utility is hampered by its dose-limiting toxicities, including nephrotoxicity, ototoxicity, neurotoxicity, and myelosuppression. This review aims to provide a comprehensive overview of cisplatin toxicity, encompassing its underlying mechanisms, risk factors, and emerging therapeutic strategies. The mechanisms of cisplatin toxicity are multifactorial and involve oxidative stress, inflammation, DNA damage, and cellular apoptosis. Various risk factors contribute to the interindividual variability in susceptibility to cisplatin toxicity. The risk of developing cisplatin-induced toxicity could be related to pre-existing conditions, including kidney disease, hearing impairment, neuropathy, impaired liver function, and other comorbidities. Additionally, this review highlights the emerging therapeutic strategies that could be applied to minimize cisplatin-induced toxicities and aid in optimizing cisplatin treatment regimens, improving patient outcomes, and enhancing the overall quality of cancer care.
Collapse
Affiliation(s)
- Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, 51452, Saudi Arabia.
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia; Central Laboratory, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Damanhour University, Egypt
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Salama A Salama
- Department of Zoology, Faculty of Science, Damanhour University, Egypt; Department of Biology, College of Science, Jazan University, Jazan 45142, Saudi Arabia
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| |
Collapse
|
9
|
Zhang M, Chen G, Chen Y, Sui Y, Zhang Y, Yang W, Yu X. Synthesis, biological activities and mechanism studies of 1,3,4-oxadiazole analogues of petiolide A as anticancer agents. Mol Divers 2024; 28:3713-3737. [PMID: 38300352 DOI: 10.1007/s11030-023-10773-w] [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: 10/12/2023] [Accepted: 11/17/2023] [Indexed: 02/02/2024]
Abstract
In order to develop new natural product-based anticancer agents, a series of 1,3,4-oxadiazole analogues based on petiolide A were prepared and evaluated for their anticancer activities by MTT method. The structures of all analogues were characterized by various spectral analyses, and B9 was further confirmed by X-ray crystallography. Among all the synthesized compounds, B1 displayed the most promising growth inhibitory effect on colon cancer cells (HCT116) with the IC50 value of 8.53 μM. Flow cytometric analysis exhibited that B1 arrested the cell cycle at G2 phase and induced apoptosis. Additionally, network pharmacology analysis calculated that B1 might target several key proteins, including AKT serine/threonine kinase 1 (AKT1), SRC proto-oncogene, non-receptor tyrosine kinase (SRC) and epidermal growth factor receptor (EGFR). Furthermore, molecular docking study indicated that B1 had potentially high binding affinity to these three target proteins. Given these results, analogue B1 could be deeply developed as potential anticancer agents.
Collapse
Affiliation(s)
- Minjie Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Guifen Chen
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yafang Chen
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yi Sui
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
- Guizhou Joint Laboratory for International Cooperation in Ethnic Medicine (Ministry of Education), Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Wude Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
- Guizhou Joint Laboratory for International Cooperation in Ethnic Medicine (Ministry of Education), Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Xiang Yu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
- Guizhou Joint Laboratory for International Cooperation in Ethnic Medicine (Ministry of Education), Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| |
Collapse
|
10
|
König P, Eichhorn JM, Suparman E, Bückreiß N, Cinatl J, Michaelis M, Bendas G. SLFN11 and ATR as targets for overcoming cisplatin resistance in ovarian cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167448. [PMID: 39117290 DOI: 10.1016/j.bbadis.2024.167448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
The levels and activities of the DNA/RNA helicase schlafen11 (SLFN11) and the serine/threonine-protein kinase ataxia telangiectasia and Rad3-related protein (ATR) may determine cancer cell sensitivity to DNA damaging agents, including platinum drugs. Here, we studied the roles of SLFN11 and ATR in cisplatin resistance of ovarian cancer using cell lines displaying acquired or intrinsic cisplatin resistance. W1CR, the cisplatin-resistant subline of W1 ovarian cancer cells, displayed reduced SLFN11 levels. HDAC inhibition using entinostat returned an epigenetic downregulation of SLFN11 in W1CR cells, caused SLFN11 re-expression and re-sensitized these cells to cisplatin. Moreover, entinostat also sensitized intrinsically resistant EFO21 ovarian cancer cells to cisplatin by upregulating SLFN11. However, SLFN11 was not involved in cisplatin resistance in all other cell models. Thus, SLFN11 expression is not a general cisplatin resistance marker in ovarian cancer. In contrast, inhibition of the DNA damage repair master regulator ATR using sub-toxic concentrations of elimusertib sensitized parental cell lines as well as intrinsically resistant EFO21 cells to cisplatin, and fully reversed acquired cisplatin resistance in cisplatin-adapted sublines W1CR, A2780cis, and KuramochirCDDP2000. Mechanisms underlying ATR-mediated cisplatin resistance differed between the cell lines and included CHK1/WEE1 signaling and induction of homologous recombination. In conclusion, SLFN11 and ATR are involved in ovarian cancer cisplatin resistance. Although our data identify ATR as key target for tackling cisplatin resistance in ovarian cancer, future studies are needed to identify biomarkers that indicate, which individual ovarian cancers benefit from SLFN11 re-activation and/or ATR inhibition.
Collapse
Affiliation(s)
- Philipp König
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany
| | | | - Eloy Suparman
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany
| | - Nico Bückreiß
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany
| | - Jindrich Cinatl
- Institute of Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany; Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60528 Frankfurt am Main, Germany
| | - Martin Michaelis
- Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60528 Frankfurt am Main, Germany; School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, Kent CT2 7NJ, UK
| | - Gerd Bendas
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany.
| |
Collapse
|
11
|
Khamto N, Utama K, Chawapun P, Siriphong S, Tateing S, Duangdesh S, Sangthong P, Chomsri NO, Meepowpan P. Discovery of amino acid-conjugated dimethylcardamonin analogues as potent anti-cervical cancer agents on SiHa cells targeting p53 signalling pathway. Biomed Pharmacother 2024; 181:117705. [PMID: 39586137 DOI: 10.1016/j.biopha.2024.117705] [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: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
DMC (1) is a phytochemical found in the seeds of Syzygium nervosum, exhibiting anticancer activity in various cells through multiple pathways. Herein, the bioactivity of DMC (1) was enhanced by chemical modification through esterification, attaching fatty acid and amino acid moieties to yield 27 semi-synthetic derivatives. These compounds were evaluated for their in vitro cytotoxicity against three main types of cervical cancer cells, including SiHa, HeLa, and C-33A. As a result, the amino acid DMC derivative, 4´-(L-tyrosinyloxy)-DMC (7j), exhibited potent cytotoxicity against SiHa cells, which was approximately two-fold greater than that of 1. Further investigation into the mechanism of action of 7j was conducted, revealing its ability to induce cell cycle arrest and apoptosis. Gene expression analysis showed the downregulation of CDK2 and upregulation of the BAX/BCL2 ratio. Atomistic insight was studied on HPV 16 E6 via molecular dynamics simulation, revealing key interactions between tyrosinyl portion and C51 residue.
Collapse
Affiliation(s)
- Nopawit Khamto
- Department of Chemistry, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; Multidisciplinary and Interdisciplinary School, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand
| | - Kraikrit Utama
- Office of Research Administration, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand
| | - Pornthip Chawapun
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand
| | - Sadanon Siriphong
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand
| | - Suriya Tateing
- Department of Plant and Soil Sciences, Faculty of Agriculture, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand
| | - Sarocha Duangdesh
- Department of Chemistry, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; Multidisciplinary and Interdisciplinary School, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand
| | - Padchanee Sangthong
- Department of Chemistry, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand
| | - Ni-Orn Chomsri
- Agricultural Technology Research Institute (ATRI), Rajamangala University of Technology Lanna, 202 Pichai District, Lampang 52100, Thailand
| | - Puttinan Meepowpan
- Department of Chemistry, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; Center of Excellence in Materials Science and Technology, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand.
| |
Collapse
|
12
|
Scanlon E, Lavery A, Albraikat M, Stevenson L, Kennedy C, Byrne R, Walker A, Mullan-Young B, McManus DT, Virdee PS, Elhussein L, Turbitt J, Collinson D, Miedzybrodzka Z, Van Schaeybroeck S, McQuaid S, James JA, Craig SG, Blayney JK, Petty RD, Harkin DP, Kennedy RD, Eatock MM, Middleton MR, Thomas A, Turkington RC. Immune microenvironment modulation following neoadjuvant therapy for oesophageal adenocarcinoma: a translational analysis of the DEBIOC clinical trial. ESMO Open 2024; 9:103930. [PMID: 39395265 PMCID: PMC11693431 DOI: 10.1016/j.esmoop.2024.103930] [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: 05/23/2024] [Revised: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND The Dual Erb B Inhibition in Oesophago-gastric Cancer (DEBIOC) trial reported an acceptable safety profile for neoadjuvant oxaliplatin and capecitabine (Xelox) ± AZD8931 in oesophageal adenocarcinoma (OAC) but limited efficacy. We evaluated the impact of neoadjuvant Xelox ± AZD8931, a novel small-molecule inhibitor with equipotent activity against epidermal growth factor receptor (EGFR), human epidermal growth factor receptor (HER)2 and HER3, on biological pathways using a unique software-driven solution. PATIENTS AND METHODS Transcriptomic profiles from 25 pre-treatment formalin-fixed paraffin-embedded OAC biopsies and 18 matched resection specimens, treated with Xelox + AZD8931 (n = 16) and Xelox alone (n = 9), were analysed using the Almac claraT total mRNA report analysing 92 gene signatures, 100 unique single-gene drug targets and 7337 single genes across 10 hallmarks of cancer. Gene-set enrichment analysis (GSEA) was utilised to investigate pathways governing pathological response. Tumour-infiltrating lymphocytes (TILs) were assessed digitally using the QuPath software. RESULTS Hierarchical clustering identified three molecular subgroups classified by activation of innate immune signalling. The immune-high subgroup was associated with HER2 positivity, increased pathological response and a marked reduction in immune signalling and TILs following neoadjuvant therapy. The immune-low cluster was predominantly HER2/EGFR-negative, and EGFR positivity was associated with the immune-mixed subgroup. Treatment with neoadjuvant therapy induced common resistance mechanisms, such as angiogenesis and epithelial-mesenchymal transition signalling, and a reduction in DNA repair signatures. Addition of AZD8931 was associated with reduction of expression of EGFR, HER2 and AKT pathways and also promoted an immunosuppressive microenvironment. GSEA showed that patients with a pathological response to treatment had increased immune signalling, whereas non-responders to neoadjuvant therapy were enriched for nucleotide repair and cellular growth through the action of E2F transcription factors. CONCLUSION OAC may be subdivided into three immune-related subgroups which undergo modulation in response to neoadjuvant therapy with marked suppression of the immune microenvironment in HER2-positive/immune-high tumours.
Collapse
Affiliation(s)
- E Scanlon
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - A Lavery
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - M Albraikat
- Precision Medicine Centre of Excellence, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - L Stevenson
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - C Kennedy
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - R Byrne
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - A Walker
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - B Mullan-Young
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - D T McManus
- Department of Pathology, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast
| | - P S Virdee
- Centre for Statistics in Medicine, University of Oxford, Oxford
| | - L Elhussein
- Centre for Statistics in Medicine, University of Oxford, Oxford
| | - J Turbitt
- Medical Genetics, School of Medicine, Medical Sciences, Nutrition and Dentistry, University of Aberdeen, Aberdeen
| | - D Collinson
- Medical Genetics, School of Medicine, Medical Sciences, Nutrition and Dentistry, University of Aberdeen, Aberdeen
| | - Z Miedzybrodzka
- Medical Genetics, School of Medicine, Medical Sciences, Nutrition and Dentistry, University of Aberdeen, Aberdeen
| | - S Van Schaeybroeck
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - S McQuaid
- Precision Medicine Centre of Excellence, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Department of Pathology, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast; Northern Ireland Biobank, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - J A James
- Precision Medicine Centre of Excellence, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Department of Pathology, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast; Northern Ireland Biobank, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - S G Craig
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - J K Blayney
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - R D Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and School of Medicine, University of Dundee, Dundee
| | - D P Harkin
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Almac Diagnostic Services Ltd, Craigavon
| | - R D Kennedy
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Almac Diagnostic Services Ltd, Craigavon
| | - M M Eatock
- Northern Ireland Cancer Centre, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast
| | - M R Middleton
- Department of Oncology, University of Oxford, Oxford
| | - A Thomas
- University of Leicester, Leicester, UK
| | - R C Turkington
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast.
| |
Collapse
|
13
|
Khan SH, Anees M, Zofair SFF, Rasool F, Khan MA, Moin S, Younus H. Fucoidan based polymeric nanoparticles encapsulating epirubicin: A novel and effective chemotherapeutic formulation against colorectal cancer. Int J Pharm 2024; 664:124622. [PMID: 39197799 DOI: 10.1016/j.ijpharm.2024.124622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Colorectal cancer (CRC) is one of the most common and challenging malignancy that needs some effective and safer chemotherapeutic agents for the treatment. In this study, anticancer agent epirubicin (Epi) was loaded in polymeric polyethylene glycol-polylactic acid-nanoparticles (mPEG-PLA-NPs) coated with a marine anti-cancer non-toxic polysaccharide fucoidan (FC), to achieve a synergistic activity against CRC. The characterization of the NPs revealed that they were spherical, monodispersed, stable, with a negative zeta potential, and exhibited good biocompatibility and controlled release. In vitro anti-cancer activity of the NPs on HCT116 cell line was found to be promising, and corroborated well with in vivo studies involving BALB/C mice injected with C26 murine cancer cells. The outcome of MTT assay demonstrated that IC50 value of free Epi was 3.72 µM, and that of non-coated and coated Epi nano-formulations was 33.67 and 10.19 µM, respectively. Higher tumor regression, better survival and reduced off-side cardiotoxicity were observed when this novel NPs formulation was used to treat tumor-bearing mice. Free FC and Epi treated mice showed 37.73 % and 61.49 % regression in tumor size, whereas there was 79.76 % and 90.34 % tumor regression in mice treated with non-coated Epi NPs and coated Epi NPs, respectively. Therefore, mPEG-PLA-FC-Epi-NPs hold a potential to be used as an effective chemotherapeutic formulation against CRC, since it exhibited better efficacy and lower toxicity.
Collapse
Affiliation(s)
- Shaheer Hasan Khan
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Mohd Anees
- Centre for Biomedical Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Syeda Fauzia Farheen Zofair
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Fayyaz Rasool
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Shagufta Moin
- Department of Biochemistry, J.N.M.C., Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Hina Younus
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India.
| |
Collapse
|
14
|
Zhang J, Chen Y, Gong X, Yang Y, Gu Y, Huang L, Fu J, Zhao M, Huang Y, Li L, Liu W, Wan Y, He X, Ma Z, Zhao W, Zhang M, Tang T, Wang Y, Thiery JP, Zheng X, Chen L. GATA factor TRPS1, a new DNA repair protein, cooperates with reversible PARylation to promote chemoresistance in patients with breast cancer. J Biol Chem 2024; 300:107780. [PMID: 39276941 PMCID: PMC11490888 DOI: 10.1016/j.jbc.2024.107780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
Resistance to DNA-damaging agents is a major unsolved challenge for breast cancer patients undergoing chemotherapy. Here, we show that elevated expression of transcriptional repressor GATA binding 1 (TRPS1) is associated with lower drug sensitivity, reduced response rate, and poor prognosis in chemotherapy-treated breast cancer patients. Mechanistically, elevated TRPS1 expression promotes hyperactivity of DNA damage repair (DDR) in breast cancer cells. We provide evidence that TRPS1 dynamically localizes to DNA breaks in a Ku70-and Ku80-dependent manner and that TRPS1 is a new member of the DDR protein family. We also discover that the dynamics of TRPS1 assembly at DNA breaks is regulated by its reversible PARylation in the DDR, and that mutations of the PARylation sites on TRPS1 lead to increased sensitivity to chemotherapeutic drugs. Taken together, our findings provide new mechanistic insights into the DDR and chemoresistance in breast cancer patients and identify TRPS1 as a critical DDR protein. TRPS1 may also be considered as a target to improve chemo-sensitization strategies and, consequently, clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yatao Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xue Gong
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Yongfeng Yang
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Yun Gu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Ling Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jianfeng Fu
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Menglu Zhao
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yehong Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lulu Li
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wenzhuo Liu
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yajie Wan
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xilin He
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhifang Ma
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Weiyong Zhao
- Department of Radiation Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meng Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tao Tang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yuzhi Wang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | | | - Xiaofeng Zheng
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China.
| | - Liming Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Jiangsu Institute of Cancer Research, Jiangsu Cancer Hospital, the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Ung J, Kassai M, Tan SF, Loughran TP, Feith DJ, Cabot MC. The Drug Transporter P-Glycoprotein and Its Impact on Ceramide Metabolism-An Unconventional Ally in Cancer Treatment. Int J Mol Sci 2024; 25:9825. [PMID: 39337312 PMCID: PMC11432138 DOI: 10.3390/ijms25189825] [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: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
The tumor-suppressor sphingolipid ceramide is recognized as a key participant in the cytotoxic mechanism of action of many types of chemotherapy drugs, including anthracyclines, Vinca alkaloids, the podophyllotoxin etoposide, taxanes, and the platinum drug oxaliplatin. These drugs can activate de novo synthesis of ceramide or stimulate the production of ceramide via sphingomyelinases to limit cancer cell survival. On the contrary, dysfunctional sphingolipid metabolism, a prominent factor in cancer survival and therapy resistance, blunts the anticancer properties of ceramide-orchestrated cell death pathways, especially apoptosis. Although P-glycoprotein (P-gp) is famous for its role in chemotherapy resistance, herein, we propose alternate interpretations and discuss the capacity of this multidrug transporter as a "ceramide neutralizer", an unwelcome event, highlighting yet another facet of P-gp's versatility in drug resistance. We introduce sphingolipid metabolism and its dysfunctional regulation in cancer, present a summary of factors that contribute to chemotherapy resistance, explain how P-gp "neutralizes" ceramide by hastening its glycosylation, and consider therapeutic applications of the P-gp-ceramide connection in the treatment of cancer.
Collapse
Affiliation(s)
- Johnson Ung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA;
| | - Miki Kassai
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, The East Carolina Diabetes and Obesity Institute, Greenville, NC 27834, USA;
| | - Su-Fern Tan
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Thomas P. Loughran
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - David J. Feith
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Myles C. Cabot
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, The East Carolina Diabetes and Obesity Institute, Greenville, NC 27834, USA;
| |
Collapse
|
16
|
Makk-Merczel K, Varga D, Hajdinák P, Szarka A. The interlacing anticancer effect of pharmacologic ascorbate, chloroquine, and resveratrol. Biofactors 2024; 50:980-996. [PMID: 38488303 DOI: 10.1002/biof.2050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/23/2024] [Indexed: 10/04/2024]
Abstract
Currently, a diagnosis with KRAS mutant pancreatic ductal adenocarcinoma (PDAC) means a death warrant, so finding efficient therapeutic options is a pressing issue. Here, we presented that pharmacologic ascorbate, chloroquine and resveratrol co-treatment exerted a synergistic cytotoxic effect on PDAC cell lines. The observed synergistic cytotoxicity was a general feature in all investigated cancer cell lines independent of the KRAS mutational status and seems to be independent of the autophagy inhibitory effect of chloroquine. Furthermore, it seems that apoptosis and necroptosis are also not likely to play any role in the cytotoxicity of chloroquine. Both pharmacologic ascorbate and resveratrol caused double-strand DNA breaks accompanied by cell cycle arrest. It seems resveratrol-induced cytotoxicity is independent of reactive oxygen species (ROS) generation and accompanied by a significant elevation of caspase-3/7 activity, while pharmacologic ascorbate-induced cytotoxicity shows strong ROS dependence but proved to be caspase-independent. Our results are particularly important since ascorbate and resveratrol are natural compounds without significant harmful effects on normal cells, and chloroquine is a known antimalarial drug that can easily be repurposed.
Collapse
Affiliation(s)
- Kinga Makk-Merczel
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
- Biotechnology Model Laboratory, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Dóra Varga
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
- Biotechnology Model Laboratory, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Péter Hajdinák
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
- Biotechnology Model Laboratory, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - András Szarka
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
- Biotechnology Model Laboratory, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| |
Collapse
|
17
|
Mukkavilli V, Ramakrishnan G, Gujjula KR, S B, Chamarthy S, Mekala JR. Molecular Understanding and Pharmacological Potency of Plant-Derived Compounds in Colorectal Cancer (CRC): A Critical Analysis and Future Perspectives. Cell Biochem Biophys 2024; 82:1777-1795. [PMID: 38965179 DOI: 10.1007/s12013-024-01370-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 07/06/2024]
Abstract
Colorectal cancer (CRC) is the main driver of fatality and the 3rd most often determined malignancy. Despite advances in detection and therapy, colorectal cancer (CRC) endures as the largest driver of cancer-related morbidity, and mortality. Modern habits and dietary negligence might be one of the reasons that have enhanced cancer prevalence. Thus, changes in Dietary habits will have a better impact, and help in finding a better cure for CRC. Initially, CRC was explored as a genetic event and currently, the research is focused on the epigenetic modifications of chromatin and microRNA (miRNA) in CRC cells. Natural products such as Curcumin, Resveratrol, Flavonoids, and Ellagitannins are been explored as compounds from the perspective of genetic, epigenetic, and miRNA modifications which will have future therapeutic aspects. Also, the extracts of these key players and their analogs will intervene the signaling pathway activation that involves in cancer propagation, apoptosis, cell cycle arrest, and epigenetic and miRNA modifications. Modulations of these miRNAs, and modification globally might have impact on CRC progression, and cancer tumor cell sensitivity.
Collapse
Affiliation(s)
- Vaagdevi Mukkavilli
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (Deemed to be University), Green Fields, 522502, Vaddeswram, AP, India
| | - Gnanasekaran Ramakrishnan
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (Deemed to be University), Green Fields, 522502, Vaddeswram, AP, India.
| | - Koteswara Reddy Gujjula
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (Deemed to be University), Green Fields, 522502, Vaddeswram, AP, India
| | - Balachandran S
- Dept of Chemical Engineering, Saveetha Engineering College, Saveetha Nagar Thandalam, 602105, Chennai, Tamil Nadu, India
| | - Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (Deemed to be University), Green Fields, 522502, Vaddeswram, AP, India
| | - Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (Deemed to be University), Green Fields, 522502, Vaddeswram, AP, India.
| |
Collapse
|
18
|
Lee JH. Targeting the ATM pathway in cancer: Opportunities, challenges and personalized therapeutic strategies. Cancer Treat Rev 2024; 129:102808. [PMID: 39106770 DOI: 10.1016/j.ctrv.2024.102808] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
Ataxia telangiectasia mutated (ATM) kinase plays a pivotal role in orchestrating the DNA damage response, maintaining genomic stability, and regulating various cellular processes. This review provides a comprehensive analysis of ATM's structure, activation mechanisms, and various functions in cancer development, progression, and treatment. I discuss ATM's dual nature as both a tumor suppressor and potential promoter of cancer cell survival in certain contexts. The article explores the complex signaling pathways mediated by ATM, its interactions with other DNA repair mechanisms, and its influence on cell cycle checkpoints, apoptosis, and metabolism. I examine the clinical implications of ATM alterations, including their impact on cancer predisposition, prognosis, and treatment response. The review highlights recent advances in ATM-targeted therapies, discussing ongoing clinical trials of ATM inhibitors and their potential in combination with other treatment modalities. I also address the challenges in developing effective biomarkers for ATM activity and patient selection strategies for personalized cancer therapy. Finally, I outline future research directions, emphasizing the need for refined biomarker development, optimized combination therapies, and strategies to overcome potential resistance mechanisms. This comprehensive overview underscores the critical importance of ATM in cancer biology and its emerging potential as a therapeutic target in precision oncology.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
19
|
Lee J, Kida K, Koh J, Liu H, Manyam GC, Gi YJ, Rampa DR, Multani AS, Wang J, Jayachandran G, Lee DW, Reuben JM, Sahin A, Huo L, Tripathy D, Im SA, Ueno NT. The DNA repair pathway as a therapeutic target to synergize with trastuzumab deruxtecan in HER2-targeted antibody-drug conjugate-resistant HER2-overexpressing breast cancer. J Exp Clin Cancer Res 2024; 43:236. [PMID: 39164784 PMCID: PMC11337831 DOI: 10.1186/s13046-024-03143-3] [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: 05/11/2023] [Accepted: 07/30/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Anti-HER2 therapies, including the HER2 antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd), have led to improved survival outcomes in patients with HER2-overexpressing (HER2+) metastatic breast cancer. However, intrinsic or acquired resistance to anti-HER2-based therapies remains a clinical challenge in these patients, as there is no standard of care following disease progression. The purpose of this study was to elucidate the mechanisms of resistance to T-DM1 and T-DXd in HER2+ BC patients and preclinical models and identify targets whose inhibition enhances the antitumor activity of T-DXd in HER2-directed ADC-resistant HER2+ breast cancer in vitro and in vivo. METHODS Targeted DNA and whole transcriptome sequencing were performed in breast cancer patient tissue samples to investigate genetic aberrations that arose after anti-HER2 therapy. We generated T-DM1 and T-DXd-resistant HER2+ breast cancer cell lines. To elucidate their resistance mechanisms and to identify potential synergistic kinase targets for enhancing the efficacy of T-DXd, we used fluorescence in situ hybridization, droplet digital PCR, Western blotting, whole-genome sequencing, cDNA microarray, and synthetic lethal kinome RNA interference screening. In addition, cell viability, colony formation, and xenograft assays were used to determine the synergistic antitumor effect of T-DXd combinations. RESULTS We found reduced HER2 expression in patients and amplified DNA repair-related genes in patients after anti-HER2 therapy. Reduced ERBB2 gene amplification in HER2-directed ADC-resistant HER2+ breast cancer cell lines was through DNA damage and epigenetic mechanisms. In HER2-directed ADC-resistant HER2+ breast cancer cell lines, our non-biased RNA interference screening identified the DNA repair pathway as a potential target within the canonical pathways to enhance the efficacy of T-DXd. We validated that the combination of T-DXd with ataxia telangiectasia and Rad3-related inhibitor, elimusertib, led to significant breast cancer cell death in vitro (P < 0.01) and in vivo (P < 0.01) compared to single agents. CONCLUSIONS The DNA repair pathways contribute to HER2-directed ADC resistance. Our data justify exploring the combination treatment of T-DXd with DNA repair-targeting drugs to treat HER2-directed ADC-resistant HER2+ breast cancer in clinical trials.
Collapse
Affiliation(s)
- Jangsoon Lee
- Section of Translational Breast Cancer Research and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Biology and Therapeutics, University of Hawai'I Cancer Center, 701 Ilalo Street, Room 622, Honolulu, HI, 96813, USA.
- Present address: Cancer Biology Program, University of Hawai'I Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - Kumiko Kida
- Section of Translational Breast Cancer Research and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Present address: Department of Breast Surgical Oncology, St. Luke's International Hospital, 9-1, Akashicho, Chuouku, Tokyo, 104-8560, Japan
| | - Jiwon Koh
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Huey Liu
- Section of Translational Breast Cancer Research and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju C Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Young Jin Gi
- Section of Translational Breast Cancer Research and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology and Therapeutics, University of Hawai'I Cancer Center, 701 Ilalo Street, Room 622, Honolulu, HI, 96813, USA
| | - Dileep R Rampa
- Cancer Biology and Therapeutics, University of Hawai'I Cancer Center, 701 Ilalo Street, Room 622, Honolulu, HI, 96813, USA
| | - Asha S Multani
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gitanjali Jayachandran
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dae-Won Lee
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-Ro Jongro-Gu, Seoul, 03080, Republic of Korea
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aysegul Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Debu Tripathy
- Section of Translational Breast Cancer Research and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-Ro Jongro-Gu, Seoul, 03080, Republic of Korea.
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Biology and Therapeutics, University of Hawai'I Cancer Center, 701 Ilalo Street, Room 622, Honolulu, HI, 96813, USA.
| |
Collapse
|
20
|
Sarogni P, Frusca V, Zamborlin A, Giannini N, Menicagli M, Brancato L, Linsalata S, Di Martino F, Gonnelli A, Paiar F, Van den Bossche J, Bogers J, Voliani V. Neoadjuvant Hyperthermia Combined with Hybrid Nanoarchitectures Enhances Chemoradiotherapy Efficacy in Head and Neck Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:43272-43282. [PMID: 39126693 DOI: 10.1021/acsami.4c07393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Head and neck squamous cell carcinomas are characterized by a high incidence of recurrence, especially in patients with locally advanced disease. Standard treatment strategies can be associated with severe side effects to healthy tissues that can negatively impact the patient's quality of life. Hyperthermia (HT) is a noninvasive treatment modality that has improved the effectiveness of chemotherapy (CT) and/or radiotherapy (RT) for the management of some solid neoplasms. In this context, the association of this approach with rationally designed nanomaterials may further enhance the treatment outcome. In this study, we demonstrate the enhanced effect of neoadjuvant HT in combination with hybrid nanoarchitectures enclosing a cisplatin prodrug (NAs-CisPt) and RT. All the treatments and their combinations have been fully evaluated by employing standardized chorioallantoic membrane tumor models of HPV-negative head and neck carcinoma. An improved tumor-shrinking effect was observed by the administration of the trimodal treatment (HT/NAs-CisPt/RT), which also highlighted a significant increase in apoptosis. Our findings demonstrate that the combination of HT with nanotechnology-based CT and RT in a certain order enhances the in vivo treatment outcome. On a broader basis, this study paves the way for the next exploration of noninvasive treatment approaches for the clinical management of oral cancer based on innovative strategies.
Collapse
Affiliation(s)
- Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Valentina Frusca
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Agata Zamborlin
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- NEST-Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Noemi Giannini
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Michele Menicagli
- Fondazione Pisana per la Scienza ONLUS, via Ferruccio Giovannini 13, S. Giuliano Terme, 56017 Pisa, Italy
| | | | - Stefania Linsalata
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Fabio Di Martino
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Alessandra Gonnelli
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Fabiola Paiar
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | | | - Johannes Bogers
- ElmediX NV, Esperantolaan 4, 3001 Heverlee, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Antwerp, Belgium
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano, 4, 16148 Genoa, Italy
| |
Collapse
|
21
|
Yao Y, Zhou S, Yan Y, Fu K, Xiao S. The tripartite motif-containing 24 is a multifunctional player in human cancer. Cell Biosci 2024; 14:103. [PMID: 39160596 PMCID: PMC11334367 DOI: 10.1186/s13578-024-01289-3] [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: 04/09/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024] Open
Abstract
Tripartite motif-containing 24 (TRIM24), also known as transcriptional intermediary factor 1α (TIF1α), is the founding member of TIF1 family. Recent evidence indicates that aberrant expression of TRIM24, functions as an oncogene, is associated with poor prognosis across various cancer types. TRIM24 exhibits a multifaceted structure comprising an N-terminal TRIM region with a RING domain, B-box type 1 and type 2 domains, and a coiled-coil region, as well as a C-terminal plant-homeodomain (PHD)-bromodomain. The bromodomain serves as a 'reader' of epigenetic histone marks, regulating chromatin structure and gene expression by linking associated proteins to acetylated nucleosomal targets, thereby controlling transcription of genes. Notably, bromodomains have emerged as compelling targets for cancer therapeutic development. In addition, TRIM24 plays specialized roles as a signal transduction molecule, orchestrating various cellular signaling cascades in cancer cells. Herein, we review the recent advancements in understanding the functions of TRIM24, and demonstrate the research progress in utilizing TRIM24 as a target for cancer therapy.
Collapse
Affiliation(s)
- Yuanbing Yao
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China
| | - Sheng Zhou
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China
- Department of Ultrasound, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yue Yan
- Yanbian University Medical School, Yanji, Jilin, China
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- Center MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, 87# Xiangya Road, Changsha, 410008, Hunan, China.
| | - Shuai Xiao
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, 69# Chuanshan Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
22
|
Djordjevic Aleksic J, Kolarević S, Jovanović Marić J, Kračun-Kolarević M, Žegura B, Štern A, Sladić D, Novaković I, Vuković-Gačić B. Influence of alkylthio and arylthio derivatives of tert-butylquinone on the induction of DNA damage in a human hepatocellular carcinoma cell line (HepG2). Toxicol In Vitro 2024; 99:105882. [PMID: 38936441 DOI: 10.1016/j.tiv.2024.105882] [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: 02/19/2024] [Revised: 05/30/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
The aim of this study was to investigate the effects of tert-butylquinone (TBQ) and its alkylthio and arylthio derivatives on DNA in vitro, using acellular and cellular test systems. Direct interaction with DNA was studied using the plasmid pUC19. Cytotoxic (MTS assay) and genotoxic (comet assay and γH2AX focus assays) effects, and their influence on the cell cycle were studied in the HepG2 cell line. Our results show that TBQ and its derivatives did not directly interact with DNA. The strongest cytotoxic effect on the HepG2 cells was observed for the derivative 2-tert-butyl-5,6-(ethylenedithio)-1,4-benzoquinone (IC50 64.68 and 55.64 μM at 24-h and 48-h treatment, respectively). The tested derivatives did not significantly influence the cell cycle distribution in the exposed cellular populations. However, all derivatives showed a genotoxic activity stronger than that of TBQ in the comet assay, with 2-tert-butyl-5,6-(ethylenedithio)-1,4-benzoquinone producing the strongest effect. The same derivative also induced DNA double-strand breaks in the γH2AX focus assay.
Collapse
Affiliation(s)
| | - Stoimir Kolarević
- University of Belgrade, Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, Belgrade, Serbia
| | - Jovana Jovanović Marić
- University of Belgrade, Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, Belgrade, Serbia
| | - Margareta Kračun-Kolarević
- University of Belgrade, Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, Belgrade, Serbia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Alja Štern
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Dušan Sladić
- University of Belgrade, Faculty of Chemistry, Belgrade, Serbia
| | - Irena Novaković
- University of Belgrade, Institute for Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Department for Chemistry, Belgrade, Serbia
| | - Branka Vuković-Gačić
- University of Belgrade, Centre for Genotoxicology and Ecogenotoxicology, Faculty of Biology, Belgrade, Serbia
| |
Collapse
|
23
|
Petrova V, Snavely AR, Splaine J, Zhen S, Singh B, Pandey R, Chen K, Cheng A, Hermawan C, Barrett LB, Smith JA, Woolf CJ. Identification of novel neuroprotectants against vincristine-induced neurotoxicity in iPSC-derived neurons. Cell Mol Life Sci 2024; 81:315. [PMID: 39066803 PMCID: PMC11335239 DOI: 10.1007/s00018-024-05340-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/05/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of cancer chemotherapy that can often limit treatment options for cancer patients or have life-long neurodegenerative consequences that reduce the patient's quality of life. CIPN is caused by the detrimental actions of various chemotherapeutic agents on peripheral axons. Currently, there are no approved preventative measures or treatment options for CIPN, highlighting the need for the discovery of novel therapeutics and improving our understanding of disease mechanisms. In this study, we utilized human-induced pluripotent stem cell (hiPSC)-derived motor neurons as a platform to mimic axonal damage after treatment with vincristine, a chemotherapeutic used for the treatment of breast cancers, osteosarcomas, and leukemia. We screened a total of 1902 small molecules for neuroprotective properties in rescuing vincristine-induced axon growth deficits. From our primary screen, we identified 38 hit compounds that were subjected to secondary dose response screens. Six compounds showed favorable pharmacological profiles - AZD7762, A-674563, Blebbistatin, Glesatinib, KW-2449, and Pelitinib, all novel neuroprotectants against vincristine toxicity to neurons. In addition, four of these six compounds also showed efficacy against vincristine-induced growth arrest in human iPSC-derived sensory neurons. In this study, we utilized high-throughput screening of a large library of compounds in a therapeutically relevant assay. We identified several novel compounds that are efficacious in protecting different neuronal subtypes from the toxicity induced by a common chemotherapeutic agent, vincristine which could have therapeutic potential in the clinic.
Collapse
Affiliation(s)
- Veselina Petrova
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew R Snavely
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jennifer Splaine
- ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Shannon Zhen
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Bhagat Singh
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Roshan Pandey
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Kuchuan Chen
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Anya Cheng
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Crystal Hermawan
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Lee B Barrett
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jennifer A Smith
- ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
- F.M. Kirby Neurobiology Center, Center for Life Science, 3 Blackfan Circle, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Petrova V, Snavely AR, Splaine J, Zhen S, Singh B, Pandey R, Chen K, Cheng A, Hermawan C, Barrett LB, Smith JA, Woolf C. Identification of novel neuroprotectants against vincristine-induced neurotoxicity in iPSC-derived neurons. RESEARCH SQUARE 2024:rs.3.rs-4545853. [PMID: 39011110 PMCID: PMC11247920 DOI: 10.21203/rs.3.rs-4545853/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of cancer chemotherapy that can often limit treatment options for cancer patients or have life-long neurodegenerative consequences that reduce the patient's quality of life. CIPN is caused by the detrimental actions of various chemotherapeutic agents on peripheral axons. Currently, there are no approved preventative measures or treatment options for CIPN, highlighting the need for the discovery of novel therapeutics and improving our understanding of disease mechanisms. In this study, we utilized human-induced pluripotent stem cell (hiPSC)-derived motor neurons as a platform to mimic axonal damage after treatment with vincristine, a chemotherapeutic used for the treatment of breast cancers, osteosarcomas, and leukemia. We screened a total of 1902 small molecules for neuroprotective properties in rescuing vincristine-induced axon growth deficits. From our primary screen, we identified 38 hit compounds that were subjected to secondary dose response screens. Six compounds showed favorable pharmacological profiles - AZD7762, A-674563, Blebbistatin, Glesatinib, KW-2449, and Pelitinib, all novel neuroprotectants against vincristine toxicity to neurons. In addition, four of these six compounds also showed efficacy against vincristine-induced growth arrest in human iPSC-derived sensory neurons. In this study, we utilized high-throughput screening of a large library of compounds in a therapeutically relevant assay. We identified several novel compounds that are efficacious in protecting different neuronal subtypes from the toxicity induced by a common chemotherapeutic agent, vincristine which could have therapeutic potential in the clinic.
Collapse
Affiliation(s)
| | | | | | - Shannon Zhen
- Boston Childrens Hospital: Boston Children's Hospital
| | - Bhagat Singh
- Boston Childrens Hospital: Boston Children's Hospital
| | | | | | - Anya Cheng
- Boston Childrens Hospital: Boston Children's Hospital
| | | | | | - Jennifer A Smith
- Harvard Medical School Center for Blood Research: Harvard Medical School
| | | |
Collapse
|
25
|
He X, Liang D, Zhou J, Li K, Xie B, Liang C, Liu C, Chen Z, Chen X, Long A, Zhuo S, Su X, Luo Y, Chen W, Zhao F, Jiang X. Nucleus-targeting DNase I self-assembly delivery system guided by pirarubicin for programmed multi-drugs release and combined anticancer therapy. Int J Biol Macromol 2024; 267:131514. [PMID: 38608986 DOI: 10.1016/j.ijbiomac.2024.131514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
The cell nucleus serves as the pivotal command center of living cells, and delivering therapeutic agents directly into the nucleus can result in highly efficient anti-tumor eradication of cancer cells. However, nucleus-targeting drug delivery is very difficult due to the presence of numerous biological barriers. Here, three antitumor drugs (DNase I, ICG: indocyanine green, and THP: pirarubicin) were sequentially triggered protein self-assembly to produce a nucleus-targeting and programmed responsive multi-drugs delivery system (DIT). DIT consisted of uniform spherical particles with a size of 282 ± 7.7 nm. The acidic microenvironment of tumors and near-infrared light could successively trigger DIT for the programmed release of three drugs, enabling targeted delivery to the tumor. THP served as a nucleus-guiding molecule and a chemotherapy drug. Through THP-guided DIT, DNase I was successfully delivered to the nucleus of tumor cells and killed them by degrading their DNA. Tumor acidic microenvironment had the ability to induce DIT, leading to the aggregation of sufficient ICG in the tumor tissues. This provided an opportunity for the photothermal therapy of ICG. Hence, three drugs were cleverly combined using a simple method to achieve multi-drugs targeted delivery and highly effective combined anticancer therapy.
Collapse
Affiliation(s)
- Xuan He
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Dan Liang
- Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Jun Zhou
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Kangjing Li
- Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Beibei Xie
- Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Chunyun Liang
- Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Cong Liu
- Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhiyong Chen
- Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xinxin Chen
- Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Ao Long
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Clinical Laboratory Medicine Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Shufang Zhuo
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Clinical Laboratory Medicine Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoping Su
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Ying Luo
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Wenxia Chen
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Conservative Dentistry & Endodontics Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Fengfeng Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| | - Xinglu Jiang
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China; Clinical Laboratory Medicine Department, College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
26
|
Burris HA, Berlin J, Arkenau T, Cote GM, Lolkema MP, Ferrer-Playan J, Kalapur A, Bolleddula J, Locatelli G, Goddemeier T, Gounaris I, de Bono J. A phase I study of ATR inhibitor gartisertib (M4344) as a single agent and in combination with carboplatin in patients with advanced solid tumours. Br J Cancer 2024; 130:1131-1140. [PMID: 38287179 PMCID: PMC10991509 DOI: 10.1038/s41416-023-02436-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Gartisertib is an oral inhibitor of ataxia telangiectasia and Rad3-related protein (ATR), a key kinase of the DNA damage response. We aimed to determine the safety and tolerability of gartisertib ± carboplatin in patients with advanced solid tumours. METHODS This phase I open-label, multicenter, first-in-human study comprised four gartisertib cohorts: A (dose escalation [DE]; Q2W); A2 (DE; QD/BID); B1 (DE+carboplatin); and C (biomarker-selected patients). RESULTS Overall, 97 patients were enroled into cohorts A (n = 42), A2 (n = 26), B1 (n = 16) and C (n = 13). The maximum tolerated dose and recommended phase II dose (RP2D) were not declared for cohorts A or B1. In cohort A2, the RP2D for gartisertib was determined as 250 mg QD. Gartisertib was generally well-tolerated; however, unexpected increased blood bilirubin in all study cohorts precluded further DE. Investigations showed that gartisertib and its metabolite M26 inhibit UGT1A1-mediated bilirubin glucuronidation in human but not dog or rat liver microsomes. Prolonged partial response (n = 1 [cohort B1]) and stable disease >6 months (n = 3) did not appear to be associated with biomarker status. Exposure generally increased dose-dependently without accumulation. CONCLUSION Gartisertib was generally well-tolerated at lower doses; however, unexpected liver toxicity prevented further DE, potentially limiting antitumour activity. Gartisertib development was subsequently discontinued. CLINICALTRIALS GOV: NCT02278250.
Collapse
Affiliation(s)
| | - Jordan Berlin
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | | | - Gregory M Cote
- Division of Hematology and Oncology, Mass General Cancer Center, Boston, MA, USA
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Utrecht, Netherlands
- Amgen Inc., Thousand Oaks, CA, USA
| | - Jordi Ferrer-Playan
- Global Clinical Development, Ares Trading SA, an affiliate of Merck KGaA, Eysins, Switzerland
| | - Anup Kalapur
- Global Patient Safety Oncology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Jayaprakasam Bolleddula
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., an affiliate of Merck KGaA, Billerica, MA, USA
| | | | | | - Ioannis Gounaris
- Global Clinical Development, Merck Serono Ltd., an affiliate of Merck KGaA, Feltham, UK
| | - Johann de Bono
- Division of Clinical Studies, Institute of Cancer Research, London, UK
- Royal Marsden, Hospital, London, UK
| |
Collapse
|
27
|
Badalamenti N, Maggio A, Fontana G, Bruno M, Lauricella M, D’Anneo A. Synthetic Derivatives of Natural ent-Kaurane Atractyligenin Disclose Anticancer Properties in Colon Cancer Cells, Triggering Apoptotic Cell Demise. Int J Mol Sci 2024; 25:3925. [PMID: 38612735 PMCID: PMC11011390 DOI: 10.3390/ijms25073925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
The antitumor activity of different ent-kaurane diterpenes has been extensively studied. Several investigations have demonstrated the excellent antitumor activity of synthetic derivatives of the diterpene atractyligenin. In this research, a series of new synthetic amides and their 15,19-di-oxo analogues obtained from atractyligenin by modifying the C-2, C-15, and C-19 positions were designed in order to dispose of a set of derivatives with different substitutions at the amidic nitrogen. Using different concentrations of the obtained compounds (10-300 μM) a reduction in cell viability of HCT116 colon cancer cells was observed at 48 h of treatment. All the di-oxidized compounds were more effective than their alcoholic precursors. The di-oxidized compounds had already reduced the viability of two colon cancer cells (HCT116 and Caco-2) at 24 h when used at low doses (2.5-15 μM), while they turned out to be poorly effective in differentiated Caco-2 cells, a model of polarized enterocytes. The data reported here provide evidence that di-oxidized compounds induced apoptotic cell death, as demonstrated by the appearance of condensed and fragmented DNA in treated cells, as well as the activation of caspase-3 and fragmentation of its target PARP-1.
Collapse
Affiliation(s)
- Natale Badalamenti
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (G.F.); (M.B.); (A.D.)
- NBFC—National Biodiversity Future Center, Piazza Marina 60, 90133 Palermo, Italy
| | - Antonella Maggio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (G.F.); (M.B.); (A.D.)
- NBFC—National Biodiversity Future Center, Piazza Marina 60, 90133 Palermo, Italy
| | - Gianfranco Fontana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (G.F.); (M.B.); (A.D.)
- NBFC—National Biodiversity Future Center, Piazza Marina 60, 90133 Palermo, Italy
| | - Maurizio Bruno
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (G.F.); (M.B.); (A.D.)
- NBFC—National Biodiversity Future Center, Piazza Marina 60, 90133 Palermo, Italy
- Centro Interdipartimentale di Ricerca “Riutilizzo Bio-Based Degli Scarti da Matrici Agroalimentari” (RIVIVE), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy
| | - Marianna Lauricella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy;
| | - Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (G.F.); (M.B.); (A.D.)
| |
Collapse
|
28
|
Hwang S, Yang S, Park K, Kim B, Kim M, Shin S, Yoo A, Ahn J, Jang J, Yim YS, Seong RH, Jeong SM. Induction of Fatty Acid Oxidation Underlies DNA Damage-Induced Cell Death and Ameliorates Obesity-Driven Chemoresistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304702. [PMID: 38145969 PMCID: PMC10933680 DOI: 10.1002/advs.202304702] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/06/2023] [Indexed: 12/27/2023]
Abstract
The DNA damage response is essential for preserving genome integrity and eliminating damaged cells. Although cellular metabolism plays a central role in cell fate decision between proliferation, survival, or death, the metabolic response to DNA damage remains largely obscure. Here, this work shows that DNA damage induces fatty acid oxidation (FAO), which is required for DNA damage-induced cell death. Mechanistically, FAO induction increases cellular acetyl-CoA levels and promotes N-alpha-acetylation of caspase-2, leading to cell death. Whereas chemotherapy increases FAO related genes through peroxisome proliferator-activated receptor α (PPARα), accelerated hypoxia-inducible factor-1α stabilization by tumor cells in obese mice impedes the upregulation of FAO, which contributes to its chemoresistance. Finally, this work finds that improving FAO by PPARα activation ameliorates obesity-driven chemoresistance and enhances the outcomes of chemotherapy in obese mice. These findings reveal the shift toward FAO induction is an important metabolic response to DNA damage and may provide effective therapeutic strategies for cancer patients with obesity.
Collapse
Affiliation(s)
- Sunsook Hwang
- Department of BiochemistryInstitute for Aging and Metabolic DiseasesDepartment of Biomedicine & Health SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591South Korea
| | - Seungyeon Yang
- Department of BiochemistryInstitute for Aging and Metabolic DiseasesDepartment of Biomedicine & Health SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591South Korea
| | - Kyungsoo Park
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- School of Biological SciencesInstitute of Molecular Biology and GeneticsSeoul National UniversitySeoul08826South Korea
| | - Byungjoo Kim
- Department of BiochemistryInstitute for Aging and Metabolic DiseasesDepartment of Biomedicine & Health SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591South Korea
| | - Minjoong Kim
- Department of BiochemistryInstitute for Aging and Metabolic DiseasesDepartment of Biomedicine & Health SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591South Korea
| | - Seungmin Shin
- Department of BiochemistryInstitute for Aging and Metabolic DiseasesDepartment of Biomedicine & Health SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591South Korea
| | - Ahyoung Yoo
- Aging and Metabolism Research GroupKorea Food Research InstituteWanju‐gun55365South Korea
| | - Jiyun Ahn
- Aging and Metabolism Research GroupKorea Food Research InstituteWanju‐gun55365South Korea
- Division of Food BiotechnologyUniversity of Science and TechnologyDaejeon34113South Korea
| | - Juneil Jang
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Yeong Shin Yim
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Rho H. Seong
- School of Biological SciencesInstitute of Molecular Biology and GeneticsSeoul National UniversitySeoul08826South Korea
| | - Seung Min Jeong
- Department of BiochemistryInstitute for Aging and Metabolic DiseasesDepartment of Biomedicine & Health SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591South Korea
| |
Collapse
|
29
|
Alemi F, Poornajaf Y, Hosseini F, Vahedian V, Gharekhani M, Shoorei H, Taheri M. Interaction between lncRNAs and RNA-binding proteins (RBPs) influences DNA damage response in cancer chemoresistance. Mol Biol Rep 2024; 51:308. [PMID: 38366290 DOI: 10.1007/s11033-024-09288-w] [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: 09/13/2023] [Accepted: 01/25/2024] [Indexed: 02/18/2024]
Abstract
The DNA damage response (DDR) is a crucial cellular signaling pathway activated in response to DNA damage, including damage caused by chemotherapy. Chemoresistance, which refers to the resistance of cancer cells to the effects of chemotherapy, poses a significant challenge in cancer treatment. Understanding the relationship between DDR and chemoresistance is vital for devising strategies to overcome this resistance and improve treatment outcomes. Long non-coding RNAs (lncRNAs) are a class of RNA molecules that do not code for proteins but play important roles in various biological processes, including cancer development and chemoresistance. RNA-binding proteins (RBPs) are a group of proteins that bind to RNA molecules and regulate their functions. The interaction between lncRNAs and RBPs has been found to regulate gene expression at the post-transcriptional level, thereby influencing various cellular processes, including DDR signaling pathways. Multiple studies have demonstrated that lncRNAs can interact with RBPs to modulate the expression of genes involved in cancer chemoresistance by impacting DDR signaling pathways. Conversely, RBPs can regulate the expression and function of lncRNAs involved in DDR. Exploring these interactions can provide valuable insights for the development of innovative therapeutic approaches to overcome chemoresistance in cancer patients. This review article aims to summarize recent research on the interaction between lncRNAs and RBPs during cancer chemotherapy, with a specific focus on DDR pathways.
Collapse
Affiliation(s)
- Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Poornajaf
- Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Foroogh Hosseini
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Vahid Vahedian
- Department of Medical Clinic, Division of Hematology/Oncology and Cellular Therapy, Faculty of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Mahdi Gharekhani
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
- Rooyesh Infertility Center, Birjand University of Medical Sciences, Birjand, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Zhang S, Tamura A, Yui N. Enhanced Tumor Targeting and Antitumor Activity of Methylated β-Cyclodextrin-Threaded Polyrotaxanes by Conjugating Cyclic RGD Peptides. Biomolecules 2024; 14:223. [PMID: 38397461 PMCID: PMC10886891 DOI: 10.3390/biom14020223] [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: 01/19/2024] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
We previously reported that acid-degradable methylated β-cyclodextrins (Me-β-CDs)-threaded polyrotaxanes (Me-PRXs) can induce autophagic cell death through endoplasmic reticulum (ER) stress-related autophagy, even in apoptosis-resistant cells. Hence, Me-PRXs show great potential as anticancer therapeutics. In this study, peptide-supermolecule conjugates were designed to achieve the targeted delivery of Me-PRX to malignant tumors. Arg-Gly-Asp peptides are well-known binding motifs of integrin αvβ3, which is overexpressed on angiogenic sites and many malignant tumors. The tumor-targeted cyclic Arg-Gly-Asp (cRGD) peptide was orthogonally post-modified to Me-PRX via click chemistry. Surface plasmon resonance (SPR) results indicated that cRGD-Me-PRX strongly binds to integrin αvβ3, whereas non-targeted cyclic Arg-Ala-Glu (cRGE) peptide conjugated to Me-PRX (cRGE-Me-PRX) failed to interact with integrins αvβ3. In vitro, cRGD-Me-PRX demonstrated enhanced cellular internalization and antitumor activity in 4T1 cells than that of unmodified Me-PRX and non-targeted cRGE-Me-PRX, due to its ability to recognize integrin αvβ3. Furthermore, cRGD-Me-PRX accumulated effectively in tumors, leading to antitumor effects, and exhibited excellent biocompatibility and safety in vivo. Therefore, cRGD conjugation to enhance selectivity for integrin αvβ3-positive cancer cells is a promising design strategy for Me-PRXs in antitumor therapy.
Collapse
Affiliation(s)
| | - Atsushi Tamura
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | | |
Collapse
|
31
|
Qian H, Margaretha Plat A, Jonker A, Hoebe RA, Krawczyk P. Super-resolution GSDIM microscopy unveils distinct nanoscale characteristics of DNA repair foci under diverse genotoxic stress. DNA Repair (Amst) 2024; 134:103626. [PMID: 38232606 DOI: 10.1016/j.dnarep.2024.103626] [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: 09/07/2023] [Revised: 12/06/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024]
Abstract
DNA double-strand breaks initiate the DNA damage response (DDR), leading to the accumulation of repair proteins at break sites and the formation of the-so-called foci. Various microscopy methods, such as wide-field, confocal, electron, and super-resolution microscopy, have been used to study these structures. However, the impact of different DNA-damaging agents on their (nano)structure remains unclear. Utilising GSDIM super-resolution microscopy, here we investigated the distribution of fluorescently tagged DDR proteins (53BP1, RNF168, MDC1) and γH2AX in U2OS cells treated with γ-irradiation, etoposide, cisplatin, or hydroxyurea. Our results revealed that both foci structure and their nanoscale ultrastructure, including foci size, nanocluster characteristics, fluorophore density and localisation, can be significantly altered by different inducing agents, even ones with similar mechanisms. Furthermore, distinct behaviours of DDR proteins were observed under the same treatment. These findings have implications for cancer treatment strategies involving these agents and provide insights into the nanoscale organisation of the DDR.
Collapse
Affiliation(s)
- Haibin Qian
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Audrey Margaretha Plat
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Ard Jonker
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Ron A Hoebe
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Przemek Krawczyk
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
32
|
Mohamed SY, Elshoky HA, El-Sayed NM, Fahmy HM, Ali MA. Ameliorative effect of zinc oxide-chitosan conjugates on the anticancer activity of cisplatin: Approach for breast cancer treatment. Int J Biol Macromol 2024; 257:128597. [PMID: 38056740 DOI: 10.1016/j.ijbiomac.2023.128597] [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: 08/23/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023]
Abstract
Breast cancer is the second most prevalent cancer affecting both males and females, comprising nearly 30 % of all cancer cases. While chemotherapeutic agents, such as cisplatin (Cis), have proven successful in cancer treatment, concerns persist regarding their efficacy and the potentially dangerous side effects. Consequently, there is a crucial and ongoing need to develop approaches that minimize side effects associated with chemotherapy. In the present work, various types of nanoparticles (NPs) were synthesized and loaded with Cis. Cis was conjugated with nanocarriers such as zinc oxide (ZnO), ZnO modified with mandelic acid and graphene oxide (GO), chitosan (CS), and CS modified with ZnO and GO to enhance the selectivity of Cis towards cancer cells. Zeta potentials and particles size were assessed using electrophoretic light scattering and dynamic light scattering. NPs were characterized using transmission electron microscopy, Fourier-transform infrared spectroscopy, and X-ray diffraction. The impact of standalone Cis as well as its nanoconjugated form on the behavior of MCF-7 cell line was investigated using WST-1 cell proliferation and apoptosis/necrosis assays. Experimental findings revealed that among the various NPs tested, ZnO, and CS NPs exhibited the highest loading percentage of Cis, surpassing the loading percentages achieved with other NPs. Cytotoxicity assay showed the enhanced effect of Cis when conjugated with ZnO and CS NPs. Flow cytometry-based assays and confocal microscopy confirmed that ZnO/Cis and CS/Cis induced apoptosis. The cisplatin-nanocomplex exhibited a descending order of early apoptosis and late apoptosis in the following order: ZnO, Cis, CS, ZnO-M, CS-GO, ZnO-GO, CS-ZnO, and CS-ZnO, Cis, CS, CS-GO, ZnO-M, ZnO, ZnO-GO, respectively. None of the nanoparticle complexes displayed a significant percentage of necrotic cells, with the highest percentage reaching 4.65 % in the case of CS-GO/Cis.
Collapse
Affiliation(s)
- Salma Y Mohamed
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt.
| | - Hisham A Elshoky
- Tumor Biology Research Program, Department of Research, Children's Cancer Hospital Egypt 57357, Cairo 11441, Egypt; Nanotechnology and Advanced Materials Central Lab., Agricultural Research Center, Giza 12619, Egypt; Regional Center for Food and Feed, Agricultural Research Center, Giza 12619, Egypt.
| | - Nayera M El-Sayed
- Physics Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt.
| | - Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt.
| | - Maha A Ali
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt.
| |
Collapse
|
33
|
Deng T, Xie L, Xiaofang C, Zhang Z, Xiao Y, Peng Y, Yin L, Fu Y, Li X. ATM-Mediated translocation of RanBPM regulates DNA damage response by stabilizing p21 in non-small cell lung cancer cells. Cell Oncol (Dordr) 2024; 47:245-258. [PMID: 37676377 PMCID: PMC10899406 DOI: 10.1007/s13402-023-00866-x] [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] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
PURPOSE Platinum-based chemotherapy remains a standard-of-care for most patients with advanced non-small cell lung cancer (NSCLC). DNA damage response (DDR) induced by platinum or Etoposide activated a panel of cell cycle-regulatory proteins including p21 through p53 pathway. Previous studies have reported that RanBPM has been involved in various cellular processes such as DDR by interacting with multiple proteins. However, the underlying mechanism remains unclear. METHODS NSCLC tissue microarrays were used for assessing the expression of RanBPM by immunohistochemical staining. The roles of RanBPM in the DDR of NSCLC progression was examined in in vitro cell lines and in vivo animal models. The regulation of RanBPM on protein stability and ubiquitination levels were investigated by immunoblots and in vivo ubiquitylation assay. RESULTS The level of p21 or RanBPM is lower in NSCLC than non-malignant tissues and has a highly positive correlation. Mechanistically, RanBPM protein physically interacts with p21, and RanBPM deubiquitinates p21 by recruiting a deubiquitinase USP11 to maintain protein stability of p21. RanBPM silencing significantly decreased p21 protein level. Conversely, RanBPM overexpression led to the accumulation of endogenous p21 protein regardless of p53 status. Functionally, RanBPM regulates DDR in a p21-dependent manner. Furthermore, DNA damage significantly promoted the nuclear translocation of RanBPM protein through ATM signaling pathways. CONCLUSION RanBPM is a novel regulator of P21 protein stability, and plays a critical role in the regulation of DDR.
Collapse
Affiliation(s)
- Tanggang Deng
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China.
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Lin Xie
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chen Xiaofang
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China
| | - Zhenbin Zhang
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yugang Xiao
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuchong Peng
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Linglong Yin
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongming Fu
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiong Li
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong, China.
- Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China.
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China.
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
34
|
Zhang Y, Chen Y, Huang W, Zhou Y, Wang Y, Fu K, Zhuang W. NPAS2 dampens chemo-sensitivity of lung adenocarcinoma cells by enhancing DNA damage repair. Cell Death Dis 2024; 15:101. [PMID: 38291048 PMCID: PMC10827782 DOI: 10.1038/s41419-023-06256-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 02/01/2024]
Abstract
Chemotherapeutic agents, including cisplatin, have remained a cornerstone of lung adenocarcinoma (LUAD) treatment and continue to play an essential role in clinical practice, despite remarkable progress in therapeutic strategies. Hence, a thorough comprehension of the molecular mechanisms underlying chemotherapeutic agent resistance is paramount. Our investigation centered on the potential involvement of the NPAS2 gene in LUAD, which is highly expressed in tumors and its high expression has been associated with unfavorable overall survival rates in patients. Intriguingly, we observed that the depletion of NPAS2 in LUAD cells resulted in increased susceptibility to cisplatin treatment. Furthermore, mRNA sequencing analysis revealed that NPAS2 deficiency downregulated genes crucial to DNA repair. Additionally, NPAS2 depletion significantly impairs γH2AX accumulation, a pivotal component of the DNA damage response. Further investigation demonstrates that NPAS2 plays a crucial role in DNA double-strand breakage repair via homology-directed repair (HDR). Our inquiry into the molecular mechanisms underlying NPAS2 regulation of DDR revealed that it may enhance the stability of H2AX mRNA by binding to its mRNA, thereby upregulating the DNA damage repair pathway. In-vivo experiments further confirmed the crucial role of NPAS2 in modulating the effect of cisplatin in LUAD. Taken together, our findings suggest that NPAS2 binds to and enhances the stability of H2AX mRNA, thereby decreasing the sensitivity of tumor cells to chemotherapy by augmenting DNA damage repair.
Collapse
Affiliation(s)
- Youyu Zhang
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Department of Cardiothoracic Vascular Surgery, Zhuzhou Central Hospital, 412001, Zhuzhou, Hunan, China
| | - Yuqiao Chen
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Wentao Huang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Yuan Zhou
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Ya Wang
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Central South University, 410031, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, 410008, Changsha, Hunan, China.
| | - Wei Zhuang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Central South University, 410031, Changsha, Hunan, China.
| |
Collapse
|
35
|
Kowalik M, Masternak J, Olszewski M, Maciejewska N, Kazimierczuk K, Sitkowski J, Dąbrowska AM, Chylewska A, Makowski M. Anticancer Study on Ir III and Rh III Half-Sandwich Complexes with the Bipyridylsulfonamide Ligand. Inorg Chem 2024; 63:1296-1316. [PMID: 38174357 DOI: 10.1021/acs.inorgchem.3c03801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Organometallic half-sandwich complexes [(η5-Cp)IrCl(L)]PF6 (1) and [(η5-Cp)RhCl(L)]PF6 (2) were prepared using pentamethylcyclopentadienyl chloride dimers of iridium(III) or rhodium(III) with the 4-amino-N-(2,2'-bipyridin-5-yl)benzenesulfonamide ligand (L) and ammonium hexafluorophosphate. The crystal structures of L, 1, and 2 were analyzed in detail. The coordination reactions of the ligand with the central ions were confirmed using various spectroscopic techniques. Additionally, the interactions between sulfaligand, Ir(III), and Rh(III) complexes with carbonic anhydrase (CA), human serum albumin (HSA), and CT-DNA were investigated. The iridium(III) complex (1) did not show any antiproliferative properties against four different cancer cell lines, i.e., nonsmall cell lung cancer A549, colon cancer HCT-116, breast cancer MCF7, lymphoblastic leukemia Nalm-6, and a nonmalignant human embryonic kidney cell line HEK293, due to high binding affinity to GSH. The sulfonamide ligand (L) and rhodium(III) complex (2) were further studied. L showed competitive inhibition toward CA, while complexes 1 and 2, uncompetitive. All compounds interacted with HSA, causing a conformational change in the protein's α-helical structure, suggesting the induction of a more open conformation in HSA, reducing its biological activity. Both L and 2 were found to induce cell death through a caspase-dependent pathway. These findings position L and 2 as potential starting compounds for pharmaceutical, therapeutic, or medicinal research.
Collapse
Affiliation(s)
- Mateusz Kowalik
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Joanna Masternak
- Institute of Chemistry, Jan Kochanowski University in Kielce, Uniwersytecka 7, 25-406 Kielce, Poland
| | - Mateusz Olszewski
- Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Natalia Maciejewska
- Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Katarzyna Kazimierczuk
- Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Jerzy Sitkowski
- Institute of Organic Chemistry, Polish Academic of Science, Marcina Kasprzaka 44/52, 01-224 Warszawa, Poland
- National Medicines Institute, Chełmska 30/34, 00-725 Warszawa, Poland
| | | | - Agnieszka Chylewska
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Mariusz Makowski
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
36
|
Nagaoka K, Bai X, Liu D, Cao K, Mulla J, Ji C, Chen H, Nisar MA, Bay A, Mueller W, Hildebrand G, Gao JS, Lu S, Setoyama H, Tanaka Y, Wands JR, Huang CK. Elevated 2-oxoglutarate antagonizes DNA damage responses in cholangiocarcinoma chemotherapy through regulating aspartate beta-hydroxylase. Cancer Lett 2024; 580:216493. [PMID: 37977350 DOI: 10.1016/j.canlet.2023.216493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/22/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Cholangiocarcinoma (CCA) is resistant to systemic chemotherapies that kill malignant cells mainly through DNA damage responses (DDRs). Recent studies suggest that the involvement of 2-oxoglutarate (2-OG) dependent dioxygenases in DDRs may be associated with chemoresistance in malignancy, but how 2-OG impacts DDRs in CCA chemotherapy remains elusive. We examined serum 2-OG levels in CCA patients before receiving chemotherapy. CCA patients are classified as progressive disease (PD), partial response (PR), and stable disease (SD) after receiving chemotherapy. CCA patients classified as PD showed significantly higher serum 2-OG levels than those defined as SD and PR. Treating CCA cells with 2-OG reduced DDRs. Overexpression of full-length aspartate beta-hydroxylase (ASPH) could mimic the effects of 2-OG on DDRs, suggesting the important role of ASPH in chemoresistance. Indeed, the knockdown of ASPH improved chemotherapy in CCA cells. Targeting ASPH with a specific small molecule inhibitor also enhanced the effects of chemotherapy. Mechanistically, ASPH modulates DDRs by affecting ATM and ATR, two of the major regulators finely controlling DDRs. More importantly, targeting ASPH improved the therapeutic potential of chemotherapy in two preclinical CCA models. Our data suggested the impacts of elevated 2-OG and ASPH on chemoresistance through antagonizing DDRs. Targeting ASPH may enhance DDRs, improving chemotherapy in CCA patients.
Collapse
Affiliation(s)
- Katsuya Nagaoka
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA; Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Xuewei Bai
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Dan Liu
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin Cao
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Joud Mulla
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Chengcheng Ji
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Hongze Chen
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA
| | - Muhammad Azhar Nisar
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA
| | - Amalia Bay
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - William Mueller
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Grace Hildebrand
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Jin-Song Gao
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Hiroko Setoyama
- Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jack R Wands
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Chiung-Kuei Huang
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
37
|
Chakraborty B, Agarwal S, Kori S, Das R, Kashaw V, Iyer AK, Kashaw SK. Multiple Protein Biomarkers and Different Treatment Strategies for Colorectal Carcinoma: A Comprehensive Prospective. Curr Med Chem 2024; 31:3286-3326. [PMID: 37151060 DOI: 10.2174/0929867330666230505165031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 05/09/2023]
Abstract
In this review, we emphasized important biomarkers, pathogenesis, and newly developed therapeutic approaches in the treatment of colorectal cancer (CRC). This includes a complete description of small-molecule inhibitors, phytopharmaceuticals with antiproliferative potential, monoclonal antibodies for targeted therapy, vaccinations as immunotherapeutic agents, and many innovative strategies to intervene in the interaction of oncogenic proteins. Many factors combine to determine the clinical behavior of colorectal cancer and it is still difficult to comprehend the molecular causes of a person's vulnerability to CRC. It is also challenging to identify the causes of the tumor's onset, progression, and responsiveness or resistance to antitumor treatment. Current recommendations for targeted medications are being updated by guidelines throughout the world in light of the growing number of high-quality clinical studies. So, being concerned about the aforementioned aspects, we have tried to present a summarized pathogenic view, including a brief description of biomarkers and an update of compounds with their underlying mechanisms that are currently under various stages of clinical testing. This will help to identify gaps or shortfalls that can be addressed in upcoming colorectal cancer research.
Collapse
Affiliation(s)
- Biswadip Chakraborty
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivangi Agarwal
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivam Kori
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Ratnesh Das
- Department of Chemistry, ISF College of Pharmacy, Moga-Punjab, India
| | - Varsha Kashaw
- Sagar Institute of Pharmaceutical Sciences, Sagar (M.P.), India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan, USA
- Molecular Imaging Program, Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Sushil Kumar Kashaw
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| |
Collapse
|
38
|
Lee HM, Wright WC, Pan M, Low J, Currier D, Fang J, Singh S, Nance S, Delahunty I, Kim Y, Chapple RH, Zhang Y, Liu X, Steele JA, Qi J, Pruett-Miller SM, Easton J, Chen T, Yang J, Durbin AD, Geeleher P. A CRISPR-drug perturbational map for identifying compounds to combine with commonly used chemotherapeutics. Nat Commun 2023; 14:7332. [PMID: 37957169 PMCID: PMC10643606 DOI: 10.1038/s41467-023-43134-0] [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: 07/10/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Combination chemotherapy is crucial for successfully treating cancer. However, the enormous number of possible drug combinations means discovering safe and effective combinations remains a significant challenge. To improve this process, we conduct large-scale targeted CRISPR knockout screens in drug-treated cells, creating a genetic map of druggable genes that sensitize cells to commonly used chemotherapeutics. We prioritize neuroblastoma, the most common extracranial pediatric solid tumor, where ~50% of high-risk patients do not survive. Our screen examines all druggable gene knockouts in 18 cell lines (10 neuroblastoma, 8 others) treated with 8 widely used drugs, resulting in 94,320 unique combination-cell line perturbations, which is comparable to the largest existing drug combination screens. Using dense drug-drug rescreening, we find that the top CRISPR-nominated drug combinations are more synergistic than standard-of-care combinations, suggesting existing combinations could be improved. As proof of principle, we discover that inhibition of PRKDC, a component of the non-homologous end-joining pathway, sensitizes high-risk neuroblastoma cells to the standard-of-care drug doxorubicin in vitro and in vivo using patient-derived xenograft (PDX) models. Our findings provide a valuable resource and demonstrate the feasibility of using targeted CRISPR knockout to discover combinations with common chemotherapeutics, a methodology with application across all cancers.
Collapse
Affiliation(s)
- Hyeong-Min Lee
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - William C Wright
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Min Pan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jonathan Low
- Department of Chemical Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Duane Currier
- Department of Chemical Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jie Fang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shivendra Singh
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Stephanie Nance
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ian Delahunty
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yuna Kim
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Richard H Chapple
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yinwen Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xueying Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jacob A Steele
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jun Yang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Adam D Durbin
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Paul Geeleher
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
39
|
Abdelmaksoud NM, Abulsoud AI, Doghish AS, Abdelghany TM. From resistance to resilience: Uncovering chemotherapeutic resistance mechanisms; insights from established models. Biochim Biophys Acta Rev Cancer 2023; 1878:188993. [PMID: 37813202 DOI: 10.1016/j.bbcan.2023.188993] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
Despite the tremendous advances in cancer treatment, resistance to chemotherapeutic agents impedes higher success rates and accounts for major relapses in cancer therapy. Moreover, the resistance of cancer cells to chemotherapy is linked to low efficacy and high recurrence of cancer. To stand up against chemotherapy resistance, different models of chemotherapy resistance have been established to study various molecular mechanisms of chemotherapy resistance. Consequently, this review is going to discuss different models of induction of chemotherapy resistance, highlighting the most common mechanisms of cancer resistance against different chemotherapeutic agents, including overexpression of efflux pumps, drug inactivation, epigenetic modulation, and epithelial-mesenchymal transition. This review aims to open a new avenue for researchers to lower the resistance to the existing chemotherapeutic agents, develop new therapeutic agents with low resistance potential, and establish possible prognostic markers for chemotherapy resistance.
Collapse
Affiliation(s)
- Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt.
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11823, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11823, Egypt
| | - Tamer M Abdelghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt.
| |
Collapse
|
40
|
Konrath F, Willenbrock M, Busse D, Scheidereit C, Wolf J. A computational model of the DNA damage-induced IKK/ NF-κB pathway reveals a critical dependence on irradiation dose and PARP-1. iScience 2023; 26:107917. [PMID: 37817938 PMCID: PMC10561052 DOI: 10.1016/j.isci.2023.107917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/01/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
The activation of IKK/NF-κB by genotoxic stress is a crucial process in the DNA damage response. Due to the anti-apoptotic impact of NF-κB, it can affect cell-fate decisions upon DNA damage and therefore interfere with tumor therapy-induced cell death. Here, we developed a dynamical model describing IKK/NF-κB signaling that faithfully reproduces quantitative time course data and enables a detailed analysis of pathway regulation. The approach elucidates a pathway topology with two hubs, where the first integrates signals from two DNA damage sensors and the second forms a coherent feedforward loop. The analyses reveal a critical role of the sensor protein PARP-1 in the pathway regulation. Introducing a method for calculating the impact of changes in individual components on pathway activity in a time-resolved manner, we show how irradiation dose influences pathway activation. Our results give a mechanistic understanding relevant for the interpretation of experimental and clinical studies.
Collapse
Affiliation(s)
- Fabian Konrath
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Michael Willenbrock
- Laboratory for Signal Transduction in Tumor Cells, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Dorothea Busse
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Claus Scheidereit
- Laboratory for Signal Transduction in Tumor Cells, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Mathematics and Computer Science, Free University Berlin, Germany
| |
Collapse
|
41
|
Fang H, Ren W, Cui Q, Liang H, Yang C, Liu W, Wang X, Liu X, Shi Y, Feng J, Chen C. Integrin β4 promotes DNA damage-related drug resistance in triple-negative breast cancer via TNFAIP2/IQGAP1/RAC1. eLife 2023; 12:RP88483. [PMID: 37787041 PMCID: PMC10547475 DOI: 10.7554/elife.88483] [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] [Indexed: 10/04/2023] Open
Abstract
Anti-tumor drug resistance is a challenge for human triple-negative breast cancer (TNBC) treatment. Our previous work demonstrated that TNFAIP2 activates RAC1 to promote TNBC cell proliferation and migration. However, the mechanism by which TNFAIP2 activates RAC1 is unknown. In this study, we found that TNFAIP2 interacts with IQGAP1 and Integrin β4. Integrin β4 activates RAC1 through TNFAIP2 and IQGAP1 and confers DNA damage-related drug resistance in TNBC. These results indicate that the Integrin β4/TNFAIP2/IQGAP1/RAC1 axis provides potential therapeutic targets to overcome DNA damage-related drug resistance in TNBC.
Collapse
Affiliation(s)
- Huan Fang
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
- Kunming College of Life Sciences, University of Chinese Academy of SciencesKunming, YunnanChina
| | - Wenlong Ren
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
- School of Life Science, University of Science & Technology of ChinaHefeiChina
| | - Qiuxia Cui
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
- Affiliated Hospital of Guangdong Medical UniversityGuangdongChina
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| | - Huichun Liang
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
| | - Chuanyu Yang
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
| | - Wenjing Liu
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
| | - Xinye Wang
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
| | - Xue Liu
- Shanghai University of Medicine & Health Sciences Affiliated Sixth People’s Hospital South CampusShanghaiChina
| | - Yujie Shi
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou UniversityZhengzhouChina
| | - Jing Feng
- Shanghai University of Medicine & Health Sciences Affiliated Sixth People’s Hospital South CampusShanghaiChina
- The Second Affiliated Hospital of the Chinese University of Hong Kong (Shenzhen)ShenzhenChina
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangdong ProvinceGuangzhouChina
| | - Ceshi Chen
- Kunming Institute of Zoology, Chinese Academy of SciencesKunming, YunnanChina
- Academy of Biomedical Engineering, Kunming Medical UniversityKunmingChina
- The Third Affiliated Hospital, Kunming Medical UniversityKunmingChina
| |
Collapse
|
42
|
Patel M, Peter ME. DISE, an ancient anti-cancer mechanism that senses mutational load in cancerous cells? Oncotarget 2023; 14:839-841. [PMID: 37747368 PMCID: PMC10519245 DOI: 10.18632/oncotarget.28466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Indexed: 09/26/2023] Open
Affiliation(s)
| | - Marcus E. Peter
- Correspondence to:Marcus E. Peter, Department of Medicine, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60616, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60616, USA email
| |
Collapse
|
43
|
Phan TTT, Truong NV, Wu WG, Su YC, Hsu TS, Lin LY. Tumor suppressor p53 mediates interleukin-6 expression to enable cancer cell evasion of genotoxic stress. Cell Death Discov 2023; 9:340. [PMID: 37696858 PMCID: PMC10495329 DOI: 10.1038/s41420-023-01638-0] [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: 01/05/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
The tumor suppressor p53 primarily functions as a mediator of DNA damage-induced cell death, thereby contributing to the efficacy of genotoxic anticancer therapeutics. Here, we show, on the contrary, that cancer cells can employ genotoxic stress-induced p53 to acquire treatment resistance through the production of the pleiotropic cytokine interleukin (IL)-6. Mechanistically, DNA damage, either repairable or irreparable, activates p53 and stimulates Caspase-2-mediated cleavage of its negative regulator mouse double minute 2 (MDM2) creating a positive feedback loop that leads to elevated p53 protein accumulation. p53 transcriptionally controls the major adenosine triphosphate (ATP) release channel pannexin 1 (Panx1), which directs IL-6 induction via a mechanism dependent on the extracellular ATP-activated purinergic P2 receptors as well as their downstream intracellular calcium (iCa2+)/PI3K/Akt/NF-ĸB signaling pathway. Thus, p53 silencing impairs Panx1 and IL-6 expression and renders cancer cells sensitive to genotoxic stress. Moreover, we confirm that IL-6 hampers the effectiveness of genotoxic anticancer agents by mitigating DNA damage, driving the expression of anti-apoptotic Bcl-2 family genes, and maintaining the migratory and invasive properties of cancer cells. Analysis of patient survival and relevant factors in lung cancer and pan-cancer cohorts supports the prognostic and clinical values of Panx1 and IL-6. Notably, IL-6 secreted by cancer cells during genotoxic treatments promotes the polarization of monocytic THP-1-derived macrophages into an alternative (M2-like) phenotype that exhibits impaired anti-survival activities but enhanced pro-metastatic effects on cancer cells as compared to nonpolarized macrophages. Our study reveals the precise mechanism for genotoxic-induced IL-6 and suggests that targeting p53-mediated IL-6 may improve the responsiveness of cancer cells to genotoxic anticancer therapy.
Collapse
Affiliation(s)
- Trinh T T Phan
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Nam V Truong
- Institute of Bioinformatics and Structural Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Wen-Guey Wu
- Institute of Bioinformatics and Structural Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Yi-Chun Su
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC.
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC.
| |
Collapse
|
44
|
Pellot Ortiz KI, Rechberger JS, Nonnenbroich LF, Daniels DJ, Sarkaria JN. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023; 11:1879. [PMID: 37509518 PMCID: PMC10377337 DOI: 10.3390/biomedicines11071879] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Inhibition of the interaction between MDM2 and p53 has emerged as a promising strategy for combating cancer, including the treatment of glioblastoma (GBM). Numerous MDM2 inhibitors have been developed and are currently undergoing rigorous testing for their potential in GBM therapy. Encouraging results from studies conducted in cell culture and animal models suggest that MDM2 inhibitors could effectively treat a specific subset of GBM patients with wild-type TP53 or functional p53. Combination therapy with clinically established treatment modalities such as radiation and chemotherapy offers the potential to achieve a more profound therapeutic response. Furthermore, an increasing array of other molecularly targeted therapies are being explored in combination with MDM2 inhibitors to increase the effects of individual treatments. While some MDM2 inhibitors have progressed to early phase clinical trials in GBM, their efficacy, alone and in combination, is yet to be confirmed. In this article, we present an overview of MDM2 inhibitors currently under preclinical and clinical investigation, with a specific focus on the drugs being assessed in ongoing clinical trials for GBM patients.
Collapse
Affiliation(s)
| | - Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Leo F Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
45
|
Keshavarz M, Xie K, Bano D, Ehninger D. Aging - what it is and how to measure it. Mech Ageing Dev 2023:111837. [PMID: 37302556 DOI: 10.1016/j.mad.2023.111837] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/27/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
The current understanding of the biology of aging is largely based on research aimed at identifying factors that influence lifespan. However, lifespan as a sole proxy measure of aging has limitations because it can be influenced by specific pathologies (not generalized physiological deterioration in old age). Hence, there is a great need to discuss and design experimental approaches that are well-suited for studies targeting the biology of aging, rather than the biology of specific pathologies that restrict the lifespan of a given species. For this purpose, we here review various perspectives on aging, discuss agreement and disagreement among researchers on the definition of aging, and show that while slightly different aspects are emphasized, a widely accepted feature, shared across many definitions, is that aging is accompanied by phenotypic changes that occur in a population over the course of an average lifespan. We then discuss experimental approaches that are in line with these considerations, including multidimensional analytical frameworks as well as designs that facilitate the proper assessment of intervention effects on aging rate. The proposed framework can guide discovery approaches to aging mechanisms in all key model organisms (e.g., mouse, fish models, D. melanogaster, C. elegans) as well as in humans.
Collapse
Affiliation(s)
- Maryam Keshavarz
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
46
|
Tonkin-Reeves A, Giuliani CM, Price JT. Inhibition of autophagy; an opportunity for the treatment of cancer resistance. Front Cell Dev Biol 2023; 11:1177440. [PMID: 37363731 PMCID: PMC10290173 DOI: 10.3389/fcell.2023.1177440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
The process of macroautophagy plays a pivotal role in the degradation of long-lived, superfluous, and damaged proteins and organelles, which are later recycled for cellular use. Normal cells rely on autophagy to combat various stressors and insults to ensure survival. However, autophagy is often upregulated in cancer cells, promoting a more aggressive phenotype that allows mutated cells to evade death after exposure to therapeutic treatments. As a result, autophagy has emerged as a significant factor in therapeutic resistance across many cancer types, with underlying mechanisms such as DNA damage, cell cycle arrest, and immune evasion. This review provides a comprehensive summary of the role of autophagy in therapeutic resistance and the limitations of available autophagic inhibitors in cancer treatment. It also highlights the urgent need to explore new inhibitors that can synergize with existing therapies to achieve better patient treatment outcomes. Advancing research in this field is crucial for developing more effective treatments that can help improve the lives of cancer patients.
Collapse
Affiliation(s)
- Asha Tonkin-Reeves
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Charlett M. Giuliani
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, Melbourne, VIC, Australia
| | - John T. Price
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
47
|
Viner-Breuer R, Golan-Lev T, Benvenisty N, Goldberg M. Genome-Wide Screening in Human Embryonic Stem Cells Highlights the Hippo Signaling Pathway as Granting Synthetic Viability in ATM Deficiency. Cells 2023; 12:1503. [PMID: 37296624 PMCID: PMC10253227 DOI: 10.3390/cells12111503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
ATM depletion is associated with the multisystemic neurodegenerative syndrome ataxia-telangiectasia (A-T). The exact linkage between neurodegeneration and ATM deficiency has not been established yet, and no treatment is currently available. In this study, we aimed to identify synthetic viable genes in ATM deficiency to highlight potential targets for the treatment of neurodegeneration in A-T. We inhibited ATM kinase activity using the background of a genome-wide haploid pluripotent CRISPR/Cas9 loss-of-function library and examined which mutations confer a growth advantage on ATM-deficient cells specifically. Pathway enrichment analysis of the results revealed the Hippo signaling pathway as a major negative regulator of cellular growth upon ATM inhibition. Indeed, genetic perturbation of the Hippo pathway genes SAV1 and NF2, as well as chemical inhibition of this pathway, specifically promoted the growth of ATM-knockout cells. This effect was demonstrated in both human embryonic stem cells and neural progenitor cells. Therefore, we suggest the Hippo pathway as a candidate target for the treatment of the devastating cerebellar atrophy associated with A-T. In addition to the Hippo pathway, our work points out additional genes, such as the apoptotic regulator BAG6, as synthetic viable with ATM-deficiency. These genes may help to develop drugs for the treatment of A-T patients as well as to define biomarkers for resistance to ATM inhibition-based chemotherapies and to gain new insights into the ATM genetic network.
Collapse
Affiliation(s)
- Ruth Viner-Breuer
- The Azrieli Center for Stem Cells and Genetic Research, The Hebrew University, Givat-Ram, Jerusalem 9190401, Israel; (R.V.-B.); (T.G.-L.)
- Department of Genetics, Institute of Life Sciences, The Hebrew University, Givat-Ram, Jerusalem 9190401, Israel
| | - Tamar Golan-Lev
- The Azrieli Center for Stem Cells and Genetic Research, The Hebrew University, Givat-Ram, Jerusalem 9190401, Israel; (R.V.-B.); (T.G.-L.)
- Department of Genetics, Institute of Life Sciences, The Hebrew University, Givat-Ram, Jerusalem 9190401, Israel
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, The Hebrew University, Givat-Ram, Jerusalem 9190401, Israel; (R.V.-B.); (T.G.-L.)
- Department of Genetics, Institute of Life Sciences, The Hebrew University, Givat-Ram, Jerusalem 9190401, Israel
| | - Michal Goldberg
- Department of Genetics, Institute of Life Sciences, The Hebrew University, Givat-Ram, Jerusalem 9190401, Israel
| |
Collapse
|
48
|
Alese OB, Wu C, Chapin WJ, Ulanja MB, Zheng-Lin B, Amankwah M, Eads J. Update on Emerging Therapies for Advanced Colorectal Cancer. Am Soc Clin Oncol Educ Book 2023; 43:e389574. [PMID: 37155942 DOI: 10.1200/edbk_389574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Colorectal cancer (CRC) is the third most common malignancy worldwide. It is projected to increase by 3.2 million new cases and account for 1.6 million deaths by 2040. Mortality is largely due to limited treatment options for patients who present with advanced disease. Thus, the development of effective and tolerable therapies is crucial. Chemotherapy has been the backbone of systemic treatment of advanced CRC, but utility has been limited because of invariable resistance to therapy, narrow mechanisms of action, and unfavorable toxicity profile. Tumors that are mismatch repair-deficient have demonstrated remarkable response to immune checkpoint inhibitor therapy. However, most CRC tumors are mismatch repair-proficient and represent an unmet medical need. Although ERBB2 amplification occurs only in a few cases, it is associated with left-sided tumors and a higher incidence of brain metastasis. Numerous combinations of HER2 inhibitors have demonstrated efficacy, and antibody-drug conjugates against HER2 represent innovative strategies in this area. The KRAS protein has been classically considered undruggable. Fortunately, new agents targeting KRAS G12C mutation represent a paradigm shift in the management of affected patients and could lead the advancement in drug development for the more common KRAS mutations. Furthermore, aberrant DNA damage response is present in 15%-20% of CRCs, and emerging innovative combinations with poly (ADP-ribose) polymerase (PARP) inhibitors could improve the current therapeutic landscape. Multiple novel biomarker-driven approaches in the management of patients with advanced CRC tumors are reviewed in this article.
Collapse
Affiliation(s)
- Olatunji B Alese
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | | | - William J Chapin
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mark B Ulanja
- Christus Ochsner St Patrick Hospital, Lake Charles, LA
| | | | | | - Jennifer Eads
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
49
|
Nandi S, Mondal A, Ghosh A, Mukherjee S, Das C. Lnc-ing epigenetic mechanisms with autophagy and cancer drug resistance. Adv Cancer Res 2023; 160:133-203. [PMID: 37704287 DOI: 10.1016/bs.acr.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Long noncoding RNAs (lncRNAs) comprise a diverse class of RNA molecules that regulate various physiological processes and have been reported to be involved in several human pathologies ranging from neurodegenerative disease to cancer. Therapeutic resistance is a major hurdle for cancer treatment. Over the past decade, several studies has emerged on the role of lncRNAs in cancer drug resistance and many trials have been conducted employing them. LncRNAs also regulate different cell death pathways thereby maintaining a fine balance of cell survival and death. Autophagy is a complex cell-killing mechanism that has both cytoprotective and cytotoxic roles. Similarly, autophagy can lead to the induction of both chemosensitization and chemoresistance in cancer cells upon therapeutic intervention. Recently the role of lncRNAs in the regulation of autophagy has also surfaced. Thus, lncRNAs can be used in cancer therapeutics to alleviate the challenges of chemoresistance by targeting the autophagosomal axis. In this chapter, we discuss about the role of lncRNAs in autophagy-mediated cancer drug resistance and its implication in targeted cancer therapy.
Collapse
Affiliation(s)
- Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Aritra Ghosh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Indian Institute of Science Education and Research, Kolkata, India
| | - Shravanti Mukherjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
50
|
Bré J, Dickson AL, Read OJ, Zhang Y, McKissock FG, Mullen P, Tang P, Zickuhr GM, Czekster CM, Harrison DJ. The novel anti-cancer fluoropyrimidine NUC-3373 is a potent inhibitor of thymidylate synthase and an effective DNA-damaging agent. Cancer Chemother Pharmacol 2023; 91:401-412. [PMID: 37000221 PMCID: PMC10156769 DOI: 10.1007/s00280-023-04528-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 03/17/2023] [Indexed: 04/01/2023]
Abstract
INTRODUCTION Fluoropyrimidines, principally 5-fluorouracil (5-FU), remain a key component of chemotherapy regimens for multiple cancer types, in particular colorectal and other gastrointestinal malignancies. To overcome key limitations and pharmacologic challenges that hinder the clinical utility of 5-FU, NUC-3373, a phosphoramidate transformation of 5-fluorodeoxyuridine, was designed to improve the efficacy and safety profile as well as the administration challenges associated with 5-FU. METHODS Human colorectal cancer cell lines HCT116 and SW480 were treated with sub-IC50 doses of NUC-3373 or 5-FU. Intracellular activation was measured by LC-MS. Western blot was performed to determine binding of the active anti-cancer metabolite FdUMP to thymidylate synthase (TS) and DNA damage. RESULTS We demonstrated that NUC-3373 generates more FdUMP than 5-FU, resulting in a more potent inhibition of TS, DNA misincorporation and subsequent cell cycle arrest and DNA damage in vitro. Unlike 5-FU, the thymineless death induced by NUC-3373 was rescued by the concurrent addition of exogenous thymidine. 5-FU cytotoxicity, however, was only reversed by supplementation with uridine, a treatment used to reduce 5-FU-induced toxicities in the clinic. This is in line with our findings that 5-FU generates FUTP which is incorporated into RNA, a mechanism known to underlie the myelosuppression and gastrointestinal inflammation associated with 5-FU. CONCLUSION Taken together, these results highlight key differences between NUC-3373 and 5-FU that are driven by the anti-cancer metabolites generated. NUC-3373 is a potent inhibitor of TS that also causes DNA-directed damage. These data support the preliminary clinical evidence that suggest NUC-3373 has a favorable safety profile in patients.
Collapse
Affiliation(s)
- Jennifer Bré
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK.
- NuCana Plc, 3 Lochside Way, Edinburgh, EH12 9DT, UK.
| | - Alison L Dickson
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
- NuCana Plc, 3 Lochside Way, Edinburgh, EH12 9DT, UK
| | - Oliver J Read
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
- NuCana Plc, 3 Lochside Way, Edinburgh, EH12 9DT, UK
| | - Ying Zhang
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | | | - Peter Mullen
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - Peijun Tang
- School of Biology, University of St Andrews, North Haugh, St Andrews, KY16 9ST, UK
| | - Greice M Zickuhr
- School of Biology, University of St Andrews, North Haugh, St Andrews, KY16 9ST, UK
| | - Clarissa M Czekster
- School of Biology, University of St Andrews, North Haugh, St Andrews, KY16 9ST, UK
| | - David J Harrison
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
- NuCana Plc, 3 Lochside Way, Edinburgh, EH12 9DT, UK
| |
Collapse
|