151
|
Meng Q, Han J, Wang P, Jia C, Guan M, Zhang B, Zhao W. BMS-794833 reduces anlotinib resistance in osteosarcoma by targeting the VEGFR/Ras/CDK2 pathway. J Bone Oncol 2024; 45:100594. [PMID: 38532893 PMCID: PMC10963651 DOI: 10.1016/j.jbo.2024.100594] [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: 11/21/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Background Osteosarcoma, a tumor that originates from bone cells, has a poor prognosis and a high degree of malignancy. Anlotinib, a small-molecule multi-target tyrosine kinase inhibitor (TKI), is the first-line drug in treating osteosarcoma, especially in late-stage osteosarcoma. However, patients often develop resistance after using anlotinib for a certain period, which poses a challenge to its further clinical application. Recently, several TKIs, for instance regorafenib and cabozantinib, have showed clinical interest in treating osteosarcoma and target both vascular endothelial growth factor receptor (VEGFR) and mesenchymal epithelial transition factor (c-MET). Therefore, the identification of new TKI warrants further investigation. Methods We performed CCK8 aasays to confirm that BMS-794833 sensitization osteosarcoma cells to anlotinib. Bioinformatics analysis and rescue experiments showed that the reduce of resistance were dependent on the VEGFR/Ras/CDK2 pathway. Cell line based xenograft model were used to demonstrate that BMS-794833 and anlotinib could synergistically treat OS. Results Here, we found that BMS-794833 reduced anlotinib resistance in osteosarcoma by targeting the VEGFR/Ras/CDK2 pathway. CCK8 assay showed that BMS-794833 significantly improved the resistance of osteosarcoma cells to anlotinib. The results of rescue experiments showed that the regulatory effects of BMS-794833 on the proliferation and drug resistance of osteosarcoma cells were dependent on the VEGFR/Ras/CDK2 pathway. In addition, BMS-794833 affected the resistance of osteosarcoma cells to anlotinib through epithelial-mesenchymal transition (EMT) and apoptosis pathways. More importantly, BMS-794833 and anlotinib exerted synergistic therapeutic effects against osteosarcoma in vivo. Conclusion Altogether, this study reveals a new (VEGFR)-targeting drug that can be combined with anlotinib for the treatment of osteosarcoma, which provides an important theoretical basis for overcoming anlotinib resistance.
Collapse
Affiliation(s)
- Qingtao Meng
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, Dalian 116028, China
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Jian Han
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Peng Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Chenxu Jia
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Mingyang Guan
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Bolun Zhang
- Department of Orthopedics, Dalian NO.3 People’s Hospital, Dalian 116091, China
| | - Wenzhi Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, Dalian 116028, China
| |
Collapse
|
152
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
153
|
Khan WJ, Ali M, Hashim S, Nawaz H, Hashim SN, Safi D, Inayat A. Use of venetoclax in t(11;14) positive relapsed/refractory multiple myeloma: A systematic review. J Oncol Pharm Pract 2024; 30:552-561. [PMID: 38113108 DOI: 10.1177/10781552231218999] [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: 12/21/2023]
Abstract
BACKGROUND The plasma cell malignancy, multiple myeloma (MM), remains incurable despite advanced treatment protocols. Overexpression of Bcl-2 (an anti-apoptotic protein), in MM harboring the translocation (11;14), contributes to resistance to prior therapy. Venetoclax, a selective oral inhibitor of BCL-2 is a novel agent that shows promise as a therapeutic agent. AIMS The objective of this systematic review is to address how the use of venetoclax, alone or as a combination regimen, contributed to the treatment of patients with t(11:14) positive relapsed/refractory multiple myeloma (RRMM). DATA SOURCES This systematic review was conducted in accordance to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and was done on 5th June 2022. A literature search was conducted on PubMed and Scopus, 145 articles were screened and 10 studies were included. Risk of bias assessment was performed using the Methodological Index for Non-Randomized Studies (MINORS) criteria. DATA SUMMARY Across the studies reviewed, a total of 311 patients were identified with t(11;14) positive RRMM. The overall response rate achieved ranged between 33% and 95.5%. Furthermore, the use of venetoclax has exhibited a favorable adverse effect profile. Side effects included hematological side effects, nausea, vomiting, and diarrhea. CONCLUSION Venetoclax demonstrates promising results. When given with drugs like dexamethasone, daratumumab and carfilzomib, a synergistic effect is seen in treating translocation (11:14) positive relapsed/refractory MM. The use of venetoclax in clinical practice can potentially improve outcomes and quality of life in RRMM patients, and future research should continue to explore this promising treatment option.
Collapse
MESH Headings
- Humans
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/adverse effects
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 14/genetics
- Drug Resistance, Neoplasm
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Neoplasm Recurrence, Local/drug therapy
- Sulfonamides/therapeutic use
- Sulfonamides/administration & dosage
- Translocation, Genetic
Collapse
Affiliation(s)
- Wardah Javed Khan
- Demonstrator (Teaching Faculty) Pharmacology and Therapeutics, Northwest School of Medicine, Peshawar, Pakistan
| | - Mubeen Ali
- Demonstrator (Teaching Faculty) Pharmacology and Therapeutics, Northwest School of Medicine, Peshawar, Pakistan
| | - Sana Hashim
- Batterjee Medical College, Jeddah, Saudi Arabia
| | - Huma Nawaz
- Demonstrator (Teaching Faculty) Pharmacology and Therapeutics, Northwest School of Medicine, Peshawar, Pakistan
| | | | - Danish Safi
- Hematology and Oncology, West Virginia University Cancer Center, Morgantown, WV, USA
| | - Arslan Inayat
- Internal Medicine, HSHS St Mary's Hospital, Decatur, IL, USA
| |
Collapse
|
154
|
Gobbo M, Joy J, Guedes H, Shazib MA, Anderson C, Abdalla-Aslan R, Peechatanan K, Lajolo C, Nasir KS, Gueiros LA, Nagarajan N, Hafezi Motlagh K, Kandwal A, Rupe C, Xu Y, Ehrenpreis ED, Tonkaboni A, Epstein JB, Bossi P, Wardill HR, Graff SL. Emerging pharmacotherapy trends in preventing and managing oral mucositis induced by chemoradiotherapy and targeted agents. Expert Opin Pharmacother 2024; 25:727-742. [PMID: 38808634 DOI: 10.1080/14656566.2024.2354451] [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] [Accepted: 05/08/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION The introduction of targeted therapy and immunotherapy has tremendously changed the clinical outcomes and prognosis of cancer patients. Despite innovative pharmacological therapies and improved radiotherapy (RT) techniques, patients continue to suffer from side effects, of which oral mucositis (OM) is still the most impactful, especially for quality of life. AREAS COVERED We provide an overview of current advances in cancer pharmacotherapy and RT, in relation to their potential to cause OM, and of the less explored and more recent literature reports related to the best management of OM. We have analyzed natural/antioxidant agents, probiotics, mucosal protectants and healing coadjuvants, pharmacotherapies, immunomodulatory and anticancer agents, photobiomodulation and the impact of technology. EXPERT OPINION The discovery of more precise pathophysiologic mechanisms of CT and RT-induced OM has outlined that OM has a multifactorial origin, including direct effects, oxidative damage, upregulation of immunologic factors, and effects on oral flora. A persistent upregulated immune response, associated with factors related to patients' characteristics, may contribute to more severe and long-lasting OM. The goal is strategies to conjugate individual patient, disease, and therapy-related factors to guide OM prevention or treatment. Despite further high-quality research is warranted, the issue of prevention is paramount in future strategies.
Collapse
Affiliation(s)
- Margherita Gobbo
- Unit of Oral and Maxillofacial Surgery, Ca' Foncello Hospital, Piazzale Ospedale, Treviso, Italy
| | - Jamie Joy
- Department of Pharmacy, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Helena Guedes
- Medical Oncology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Porto, Portugal
| | - Muhammad Ali Shazib
- Workman School of Dental Medicine, High Point University, High Point, NC, USA
| | - Carryn Anderson
- Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, USA
| | - Ragda Abdalla-Aslan
- Department of Oral and Maxillofacial Surgery, Rambam Health Care Campus, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Khunthong Peechatanan
- Supportive and Palliative Care Unit, Monash Health, Clayton, VIC, Australia
- Department of Medicine, Division of Medical Oncology, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - Carlo Lajolo
- Head and Neck Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS, School of Dentistry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Khawaja Shehryar Nasir
- Department of Internal Medicine, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Luiz Alcino Gueiros
- Department of Clinic and Preventive Dentistry & Oral Medicine Unit, Health Sciences Center, Hospital das Clínicas, Federal University of Pernambuco, Recife, Brazil
| | - Nivethitha Nagarajan
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, California, USA
| | - Kimia Hafezi Motlagh
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Abhishek Kandwal
- Himalayan Institute of Medical Sciences Cancer Research Institute Swami Rama Himalayan University, Uttarakhand, India
| | - Cosimo Rupe
- Head and Neck Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS, School of Dentistry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Yuanming Xu
- Department of Diagnostic Sciences, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Eli D Ehrenpreis
- Department of Medicine, Advocate Lutheran General Hospital, Park Ridge, IL, USA
- E2Bio Life Sciences, Skokie, IL, USA
| | - Arghavan Tonkaboni
- Oral Medicine Department, School of Dentistry, Tehran University of Medical Science, Tehran, Iran
| | - Joel B Epstein
- Department of Surgery, City of Hope National Cancer Center, Duarte, CA, USA
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Hannah R Wardill
- School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Stephanie L Graff
- Lifespan Cancer Institute, Providence, RI, USA
- Legorreta Cancer Center, Brown University, Providence, RI, USA
| |
Collapse
|
155
|
Sun Y, Gong J, Li Z, Han L, Sun D. Gallbladder cancer: surgical treatment, immunotherapy, and targeted therapy. Postgrad Med 2024; 136:278-291. [PMID: 38635593 DOI: 10.1080/00325481.2024.2345585] [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: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Gallbladder cancer is a common type of biliary tract tumor. Optimal management for early stage cases typically involves radical excision as the primary treatment modality. Various surgical techniques, including laparoscopic, robotic, and navigational surgery, have demonstrated favorable clinical outcomes in radical gallbladder excision. Unfortunately, most patients are ineligible for surgical intervention because of the advanced stage of the disease upon diagnosis. Consequently, non-surgical interventions, such as chemotherapy, radiotherapy, immunotherapy, and targeted therapy, have become the mainstay of treatment for patients in advanced stages. This review focuses on elucidating various surgical techniques as well as advancements in immunotherapy and targeted therapy in the context of recent advancements in gallbladder cancer research.
Collapse
Affiliation(s)
- Yanjun Sun
- Department of General Surgery, The Armed Police Corps Hospital of Anhui, Hefei, China
| | - Junfeng Gong
- Department of General Surgery, The Armed Police Corps Hospital of Anhui, Hefei, China
| | | | - Lin Han
- Department of General Surgery, The Armed Police Corps Hospital of Anhui, Hefei, China
| | - Dengqun Sun
- Department of General Surgery, The Armed Police Corps Hospital of Anhui, Hefei, China
| |
Collapse
|
156
|
Gao F, You X, Yang L, Zou X, Sui B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int Rev Immunol 2024; 43:280-308. [PMID: 38525925 DOI: 10.1080/08830185.2024.2333275] [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: 11/05/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xiaoqing You
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Oncology, Da Qing Long Nan Hospital, Daqing, Heilongjiang Province, China
| | - Xiangni Zou
- Department of Nursing, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Bowen Sui
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
157
|
Zhang L, Zhang X, Shi Y, Ni Y, Fei J, Jin Z, Li W, Wang X, Wu N. Role and potential therapeutic value of histone methyltransferases in drug resistance mechanisms in lung cancer. Front Oncol 2024; 14:1376916. [PMID: 38525426 PMCID: PMC10957659 DOI: 10.3389/fonc.2024.1376916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/26/2024] Open
Abstract
Lung cancer, ranking second globally in both incidence and high mortality among common malignant tumors, presents a significant challenge with frequent occurrences of drug resistance despite the continuous emergence of novel therapeutic agents. This exacerbates disease progression, tumor recurrence, and ultimately leads to poor prognosis. Beyond acquired resistance due to genetic mutations, mounting evidence suggests a critical role of epigenetic mechanisms in this process. Numerous studies have indicated abnormal expression of Histone Methyltransferases (HMTs) in lung cancer, with the abnormal activation of certain HMTs closely linked to drug resistance. HMTs mediate drug tolerance in lung cancer through pathways involving alterations in cellular metabolism, upregulation of cancer stem cell-related genes, promotion of epithelial-mesenchymal transition, and enhanced migratory capabilities. The use of HMT inhibitors also opens new avenues for lung cancer treatment, and targeting HMTs may contribute to reversing drug resistance. This comprehensive review delves into the pivotal roles and molecular mechanisms of HMTs in drug resistance in lung cancer, offering a fresh perspective on therapeutic strategies. By thoroughly examining treatment approaches, it provides new insights into understanding drug resistance in lung cancer, supporting personalized treatment, fostering drug development, and propelling lung cancer therapy into novel territories.
Collapse
Affiliation(s)
- Linxiang Zhang
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xueying Zhang
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yan Shi
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuhan Ni
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jiaojiao Fei
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhixin Jin
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Wenjuan Li
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaojing Wang
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Nan Wu
- Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, The Department of Pulmonary Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
158
|
Al-Hawary SIS, Jasim SA, Hjazi A, Oghenemaro EF, Kaur I, Kumar A, Al-Ani AM, Alwaily ER, Redhee AH, Mustafa YF. Nucleic acid-based vaccine for ovarian cancer cells; bench to bedside. Cell Biochem Funct 2024; 42:e3978. [PMID: 38515237 DOI: 10.1002/cbf.3978] [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: 01/23/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024]
Abstract
Ovarian cancer continues to be a difficult medical issue that affects millions of individuals worldwide. Important platforms for cancer immunotherapy include checkpoint inhibitors, chimeric antigen receptor T cells, bispecific antibodies, cancer vaccines, and other cell-based treatments. To avoid numerous infectious illnesses, conventional vaccinations based on synthetic peptides, recombinant subunit vaccines, and live attenuated and inactivated pathogens are frequently utilized. Vaccine manufacturing processes, however, are not entirely safe and carry a significant danger of contaminating living microorganisms. As a result, the creation of substitute vaccinations is required for both viral and noninfectious illnesses, including cancer. Recently, there has been testing of nucleic acid vaccines, or NAVs, as a cancer therapeutic. Tumor antigens (TAs) are genetically encoded by DNA and mRNA vaccines, which the host uses to trigger immune responses against ovarian cancer cells that exhibit the TAs. Despite being straightforward, safe, and easy to produce, NAVs are not currently thought to be an ideal replacement for peptide vaccines. Some obstacles to this strategy include selecting the appropriate therapeutic agents (TAs), inadequate immunogenicity, and the immunosuppressive characteristic of ovarian cancer. We focus on strategies that have been employed to increase NAVs' effectiveness in the fight against ovarian cancer in this review.
Collapse
Affiliation(s)
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Anbar, Iraq
- Biotechnology Department, College of Applied Science, Fallujah University, Fallujah, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after The First President of Russia, Yekaterinburg, Russia
| | | | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
159
|
Zhou L, Lu Y, Liu W, Wang S, Wang L, Zheng P, Zi G, Liu H, Liu W, Wei S. Drug conjugates for the treatment of lung cancer: from drug discovery to clinical practice. Exp Hematol Oncol 2024; 13:26. [PMID: 38429828 PMCID: PMC10908151 DOI: 10.1186/s40164-024-00493-8] [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: 01/06/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024] Open
Abstract
A drug conjugate consists of a cytotoxic drug bound via a linker to a targeted ligand, allowing the targeted delivery of the drug to one or more tumor sites. This approach simultaneously reduces drug toxicity and increases efficacy, with a powerful combination of efficient killing and precise targeting. Antibody‒drug conjugates (ADCs) are the best-known type of drug conjugate, combining the specificity of antibodies with the cytotoxicity of chemotherapeutic drugs to reduce adverse reactions by preferentially targeting the payload to the tumor. The structure of ADCs has also provided inspiration for the development of additional drug conjugates. In recent years, drug conjugates such as ADCs, peptide‒drug conjugates (PDCs) and radionuclide drug conjugates (RDCs) have been approved by the Food and Drug Administration (FDA). The scope and application of drug conjugates have been expanding, including combination therapy and precise drug delivery, and a variety of new conjugation technology concepts have emerged. Additionally, new conjugation technology-based drugs have been developed in industry. In addition to chemotherapy, targeted therapy and immunotherapy, drug conjugate therapy has undergone continuous development and made significant progress in treating lung cancer in recent years, offering a promising strategy for the treatment of this disease. In this review, we discuss recent advances in the use of drug conjugates for lung cancer treatment, including structure-based drug design, mechanisms of action, clinical trials, and side effects. Furthermore, challenges, potential approaches and future prospects are presented.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Liu
- Department of Geriatrics, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shanglong Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guisha Zi
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| |
Collapse
|
160
|
Zhang C, Shen Q, Gao M, Li J, Pang B. The role of Cyclin Dependent Kinase Inhibitor 3 ( CDKN3) in promoting human tumors: Literature review and pan-cancer analysis. Heliyon 2024; 10:e26061. [PMID: 38380029 PMCID: PMC10877342 DOI: 10.1016/j.heliyon.2024.e26061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Background Although many experiments and clinical studies have proved the link between the expression of CDKN3 and human tumors, we have not been able to identify any bioinformatics study in which the extensive tumor-promoting effect of CDKN3 was systematically analyzed. Objective Explore the extensive tumor-promoting effects of CDKN3 and review the research progress of CDKN3 in cancer. Methods We systematically reviewed the literature on CDKN3 and tumors. We explored the potential tumor-promoting effects of CDKN3 on different tumors in the TCGA database and the GTEx database using multiple platforms and websites. We studied the expression level of CDKN3, survival, prognosis, diagnosis, genetic variation, immune infiltration, and enrichment analysis using databases such as TIMER 2.0, GEPIA2, cBioPortal, and STRING. Results We found that CDKN3 is highly expressed in most tumors. The expression of CDKN3 is closely related to the prognosis of some tumors. And CDKN3 may have diagnostic value. The conclusion of our literature review is roughly the same, but there are differences, which are worthy of further study. Moreover, CDKN3 may be related to immune cell infiltration in tumor tissues. The genetic alteration of LUAD, STAD, SARC, PCPG, and ESCA with "Amplification" as the main type. In addition, through enrichment analysis, we found that CDKN3 affects tumors mainly through the control of the cell cycle and mitosis. Conclusion CDKN3 is highly expressed in most tumor tissues and has a statistical correlation with survival prognosis. It has extensive tumor-promoting effects that may be related to mechanisms such as immune infiltration.
Collapse
Affiliation(s)
- Chuanlong Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Qian Shen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Mengqi Gao
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Junchen Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300000, China
| | - Bo Pang
- International Medical Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| |
Collapse
|
161
|
Vaswani CM, Simone J, Pavelick JL, Wu X, Tan GW, Ektesabi AM, Gupta S, Tsoporis JN, Dos Santos CC. Tiny Guides, Big Impact: Focus on the Opportunities and Challenges of miR-Based Treatments for ARDS. Int J Mol Sci 2024; 25:2812. [PMID: 38474059 DOI: 10.3390/ijms25052812] [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: 01/28/2024] [Revised: 02/24/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is characterized by lung inflammation and increased membrane permeability, which represents the leading cause of mortality in ICUs. Mechanical ventilation strategies are at the forefront of supportive approaches for ARDS. Recently, an increasing understanding of RNA biology, function, and regulation, as well as the success of RNA vaccines, has spurred enthusiasm for the emergence of novel RNA-based therapeutics. The most common types of RNA seen in development are silencing (si)RNAs, antisense oligonucleotide therapy (ASO), and messenger (m)RNAs that collectively account for 80% of the RNA therapeutics pipeline. These three RNA platforms are the most mature, with approved products and demonstrated commercial success. Most recently, miRNAs have emerged as pivotal regulators of gene expression. Their dysregulation in various clinical conditions offers insights into ARDS pathogenesis and offers the innovative possibility of using microRNAs as targeted therapy. This review synthesizes the current state of the literature to contextualize the therapeutic potential of miRNA modulation. It considers the potential for miR-based therapeutics as a nuanced approach that incorporates the complexity of ARDS pathophysiology and the multifaceted nature of miRNA interactions.
Collapse
Affiliation(s)
- Chirag M Vaswani
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Julia Simone
- Department of Medicine, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Jacqueline L Pavelick
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiao Wu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Greaton W Tan
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Amin M Ektesabi
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sahil Gupta
- Faculty of Medicine, School of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - James N Tsoporis
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Claudia C Dos Santos
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Interdepartmental Division of Critical Care, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| |
Collapse
|
162
|
Skowicki M, Hürlimann D, Tarvirdipour S, Kyropoulou M, Schoenenberger CA, Gerber-Lemaire S, Palivan CG. FAP Targeting of Photosensitizer-Loaded Polymersomes for Increased Light-Activated Cell Killing. Biomacromolecules 2024; 25:754-766. [PMID: 38267014 PMCID: PMC10865352 DOI: 10.1021/acs.biomac.3c00943] [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: 09/06/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 01/26/2024]
Abstract
As current chemo- and photodynamic cancer therapies are associated with severe side effects due to a lack of specificity and to systemic toxicity, innovative solutions in terms of targeting and controlled functionality are in high demand. Here, we present the development of a polymersome nanocarrier equipped with targeting molecules and loaded with photosensitizers for efficient uptake and light-activated cell killing. Polymersomes were self-assembled in the presence of photosensitizers from a mixture of nonfunctionalized and functionalized PDMS-b-PMOXA diblock copolymers, the latter designed for coupling with targeting ligands. By encapsulation inside the polymersomes, the photosensitizer Rose Bengal was protected, and its uptake into cells was mediated by the nanocarrier. Inhibitor of fibroblast activation protein α (FAPi), a ligand for FAP, was attached to the polymersomes' surface and improved their uptake in MCF-7 breast cancer cells expressing relatively high levels of FAP on their surface. Once internalized by MCF-7, irradiation of Rose Bengal-loaded FAPi-polymersomes generated reactive oxygen species at levels high enough to induce cell death. By combining photosensitizer encapsulation and specific targeting, polymersomes represent ideal candidates as therapeutic nanocarriers in cancer treatment.
Collapse
Affiliation(s)
- Michal Skowicki
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Dimitri Hürlimann
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Shabnam Tarvirdipour
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
| | - Myrto Kyropoulou
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Sandrine Gerber-Lemaire
- Group
for Functionalized Biomaterials, Institute of Chemical Sciences and
Engineering, Ecole Polytechnique Fédérale
de Lausanne, CH-1015 Lausanne, Switzerland
| | - Cornelia G. Palivan
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| |
Collapse
|
163
|
Mourya A, Prajapati N. Precision Deuteration in Search of Anticancer Agents: Approaches to Cancer Drug Discovery. Cancer Biother Radiopharm 2024; 39:1-18. [PMID: 37585602 DOI: 10.1089/cbr.2023.0031] [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: 08/18/2023] Open
Abstract
Cancer chemotherapy has been shifted from conventional cytotoxic drug therapy to selective and target-specific therapy after the findings about DNA changes and proteins that are responsible for cancer. A large number of newer drugs were discovered as targeted therapy for particular types of neoplastic disease. The initial discovery includes the development of the first in the category, imatinib, a Bcr-Abl tyrosine kinase inhibitor (TKI) for the treatment of chronic myelocytic leukemia in 2001. But the joy did not last for long as the drug developed a point mutation within the ABL1 kinase domain of BCR-ABL1, which subsequently led to the discovery of many other TKIs. Resistance was observed for newer TKIs a few years after their launching, but the use of TKIs in life-threatening cancer therapy is considered as far better compared with the risks of disease because of its target specificity and hence less toxicity. In search of a better anticancer agent, the physiochemical properties of the lead molecule have been modified for its efficacy toward disease and delay in the development of resistance. Deuteration in the drug molecule is one of such modifications that alter the pharmacokinetic properties, generally its metabolism, as compared with its pharmacodynamic effects. Precision deuteration in many anticancer drugs has been carried out to search for better drugs for cancer. In this review, the majority of anticancer drugs and molecules for which deuteration was applied to get better anticancer molecules were discussed. This review will provide a complete guide about the benefits of deuteration in cancer chemotherapy.
Collapse
MESH Headings
- Humans
- Drug Resistance, Neoplasm/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Imatinib Mesylate/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Drug Discovery
Collapse
Affiliation(s)
- Aman Mourya
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Navnit Prajapati
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
164
|
Nagainallur Ravichandran S, Das D, Dayananda EK, Dey A, Banerjee A, Sun-Zhang A, Zhang H, Sun XF, Pathak S. A Review on Emerging Techniques for Diagnosis of Colorectal Cancer. Cancer Invest 2024; 42:119-140. [PMID: 38404236 DOI: 10.1080/07357907.2024.2315443] [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: 06/11/2023] [Accepted: 02/02/2024] [Indexed: 02/27/2024]
Abstract
Common detection methods in practice for diagnosing colorectal cancer (CRC) are painful and invasive leading to less participation of individuals for CRC diagnosis. Whereas, improved or enhanced imaging systems and other minimally invasive techniques with shorter detection times deliver greater detail and less discomfort in individuals. Thus, this review is a summary of the diagnostic tests, ranging from the simple potential use in developing a flexible CRC treatment to the patient's potential benefits in receiving less invasive procedures and the advanced treatments that might provide a better assessment for the diagnosis of CRC and reduce the mortality related to CRC.
Collapse
Affiliation(s)
- Shruthi Nagainallur Ravichandran
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Erica Katriel Dayananda
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Amit Dey
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Alexander Sun-Zhang
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Hong Zhang
- Faculty of Medicine and Health, School of Medical Sciences, Orebro University, Örebro, Sweden
| | - Xiao-Feng Sun
- Division of Oncology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| |
Collapse
|
165
|
Qiao Y, Shi Y, Ji M, Wang Z, Bai X, Zhang K, Yin K, Zhang Y, Chen X, Zhang Y, Lu J, Zhao J, Liu K, Yuan B. Selection and identification of a prohibitin 2-binding DNA aptamer for tumor tissue imaging and targeted chemotherapy. Int J Biol Macromol 2024; 259:129002. [PMID: 38176501 DOI: 10.1016/j.ijbiomac.2023.129002] [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: 06/09/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Tumor cell-targeting molecules play a vital role in cancer diagnosis, targeted therapy, and biomarker discovery. Aptamers are emerging as novel targeting molecules with unique advantages in cancer research. In this work, we have developed several DNA aptamers through cell-based systematic evolution of ligands by exponential enrichment (Cell-SELEX). The selected SYL-6 aptamer can bind to a variety of cancer cells with high signal. Tumor tissue imaging demonstrated that SYL-6-Cy5 fluorescent probe was able to recognize multiple clinical tumor tissues but not the normal tissues, which indicates great potential of SYL-6 for clinical tumor diagnosis. Meanwhile, we identified prohibitin 2 (PHB2) as the molecular target of SYL-6 using mass spectrometry, pull-down and RNA interference assays. Moreover, SYL-6 can be used as a delivery vehicle to carry with doxorubicin (Dox) chemotherapeutic agents for antitumor targeted chemotherapy. The constructed SYL-6-Dox can not only selectively kill tumor cells in vitro, but also inhibit tumor growth with reduced side effects in vivo. This work may provide a general tumor cell-targeting molecule and a potential biomarker for cancer diagnosis and targeted therapy.
Collapse
Affiliation(s)
- Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450000, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yanli Shi
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Department of Pathology, Zhoukou Central Hospital, Zhoukou 466000, Henan, China
| | - Mengmeng Ji
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Zhaoting Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xue Bai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Kai Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Kai Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yangyang Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450000, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yueteng Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450000, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450000, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450000, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450003, Henan, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, Henan, China.
| | - Baoyin Yuan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450000, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
166
|
Dailah HG, Hommdi AA, Koriri MD, Algathlan EM, Mohan S. Potential role of immunotherapy and targeted therapy in the treatment of cancer: A contemporary nursing practice. Heliyon 2024; 10:e24559. [PMID: 38298714 PMCID: PMC10828696 DOI: 10.1016/j.heliyon.2024.e24559] [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: 04/29/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Immunotherapy and targeted therapy have emerged as promising therapeutic options for cancer patients. Immunotherapies induce a host immune response that mediates long-lived tumor destruction, while targeted therapies suppress molecular mechanisms that are important for tumor maintenance and growth. In addition, cytotoxic agents and targeted therapies regulate immune responses, which increases the chances that these therapeutic approaches may be efficiently combined with immunotherapy to ameliorate clinical outcomes. Various studies have suggested that combinations of therapies that target different stages of anti-tumor immunity may be synergistic, which can lead to potent and more prolonged responses that can achieve long-lasting tumor destruction. Nurses associated with cancer patients should have a better understanding of the immunotherapies and targeted therapies, such as their efficacy profiles, mechanisms of action, as well as management and prophylaxis of adverse events. Indeed, this knowledge will be important in establishing care for cancer patients receiving immunotherapies and targeted therapies for cancer treatment. Moreover, nurses need a better understanding regarding targeted therapies and immunotherapies to ameliorate outcomes in patients receiving these therapies, as well as management and early detection of possible adverse effects, especially adverse events associated with checkpoint inhibitors and various other therapies that control T-cell activation causing autoimmune toxicity. Nurses practice in numerous settings, such as hospitals, home healthcare agencies, radiation therapy facilities, ambulatory care clinics, and community agencies. Therefore, as compared to other members of the healthcare team, nurses often have better opportunities to develop the essential rapport in providing effective nurse-led patient education, which is important for effective therapeutic outcomes and continuance of therapy. In this article, we have particularly focused on providing a detailed overview on targeted therapies and immunotherapies used in cancer treatment, management of their associated adverse events, and the impact as well as strategies of nurse-led patient education.
Collapse
Affiliation(s)
- Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Abdullah Abdu Hommdi
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Mahdi Dafer Koriri
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Essa Mohammed Algathlan
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| |
Collapse
|
167
|
Caraban BM, Aschie M, Deacu M, Cozaru GC, Pundiche MB, Orasanu CI, Voda RI. A Narrative Review of Current Knowledge on Cutaneous Melanoma. Clin Pract 2024; 14:214-241. [PMID: 38391404 PMCID: PMC10888040 DOI: 10.3390/clinpract14010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Cutaneous melanoma is a public health problem. Efforts to reduce its incidence have failed, as it continues to increase. In recent years, many risk factors have been identified. Numerous diagnostic systems exist that greatly assist in early clinical diagnosis. The histopathological aspect illustrates the grim nature of these cancers. Currently, pathogenic pathways and the tumor microclimate are key to the development of therapeutic methods. Revolutionary therapies like targeted therapy and immune checkpoint inhibitors are starting to replace traditional therapeutic methods. Targeted therapy aims at a specific molecule in the pathogenic chain to block it, stopping cell growth and dissemination. The main function of immune checkpoint inhibitors is to boost cellular immunity in order to combat cancer cells. Unfortunately, these therapies have different rates of effectiveness and side effects, and cannot be applied to all patients. These shortcomings are the basis of increased incidence and mortality rates. This study covers all stages of the evolutionary sequence of melanoma. With all these data in front of us, we see the need for new research efforts directed at therapies that will bring greater benefits in terms of patient survival and prognosis, with fewer adverse effects.
Collapse
Affiliation(s)
- Bogdan Marian Caraban
- Clinical Department of Plastic Surgery, Microsurgery-Reconstructive, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| | - Mariana Aschie
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Academy of Medical Sciences of Romania, 030171 Bucharest, Romania
- The Romanian Academy of Scientists, 030167 Bucharest, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| | - Mariana Deacu
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Georgeta Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
- Clinical Service of Pathology, Departments of Genetics, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Mihaela Butcaru Pundiche
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Department of General Surgery, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Cristian Ionut Orasanu
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| | - Raluca Ioana Voda
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| |
Collapse
|
168
|
Al-Jumaili MMO. Transcription Silencing and CpGs Hypermethylation as Therapeutic Gene Editing in Clinical Colorectal Adenocarcinoma Repression. THE KOREAN JOURNAL OF GASTROENTEROLOGY = TAEHAN SOHWAGI HAKHOE CHI 2024; 83:6-16. [PMID: 38268163 DOI: 10.4166/kjg.2023.132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 01/26/2024]
Abstract
Background/Aims Colorectal cancer is the most common cancer in oncopathology, with an increasing incidence among the elderly during the last decade. Various genetic and environmental factors play important roles in the emergence of colorectal adenocarcinoma. Non-coding RNAs, approximately 20-22 nucleotides, are transcribed irregularly in many cancer cells and play a critical role in many metabolic pathways in clinical cancer cases. DNA methylation is a critical epigenetic alteration that controls gene expression. In the current study, transcriptional silencing and CpG hypermethylation were developed as a therapeutic gene editing strategy for the clinical repression of colorectal adenocarcinoma. Methods A human colorectal adenocarcinoma cell line (Caco2) and a normal lung fibroblast cell line (Wi38) were utilized as the paradigms in this research to examine the effect of mir155 molecule transfection and CpGs-island (CGI) methylation. Cell counting was achieved using six-well and 24-well plates before transfection using a hemocytometer. The two cell lines were transfected with the mir155 agomir and antagomir molecules. The transfection efficiency, cell viability, cell IC50, and target gene expression were measured, and CGIs-methylation was achieved by bisulfate conversion. Results The outcomes revealed the downregulation of oncogenes (AKT1 and VCAM1 genes as cancer-associated genes) and the upregulation of tumor suppressor genes (TSGs, Tp53 and KEAP1). In addition, CpG-islands methylation showed significant blocking of the oncogene promoter regions, and the switch on of TSG promoter regions was continuous. Conclusions miRNA-CGI-methylation led to the regression of Caco2 cell proliferation, suggesting the potential use of RNA silencing and DNA methylation in targeted gene therapy for colorectal cancer.
Collapse
|
169
|
Gupta D, Roy P, Sharma R, Kasana R, Rathore P, Gupta TK. Recent nanotheranostic approaches in cancer research. Clin Exp Med 2024; 24:8. [PMID: 38240834 PMCID: PMC10799106 DOI: 10.1007/s10238-023-01262-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/07/2023] [Indexed: 01/22/2024]
Abstract
Humanity is suffering from cancer which has become a root cause of untimely deaths of individuals around the globe in the recent past. Nanotheranostics integrates therapeutics and diagnostics to monitor treatment response and enhance drug efficacy and safety. We hereby propose to discuss all recent cancer imaging and diagnostic tools, the mechanism of targeting tumor cells, and current nanotheranostic platforms available for cancer. This review discusses various nanotheranostic agents and novel molecular imaging tools like MRI, CT, PET, SPEC, and PAT used for cancer diagnostics. Emphasis is given to gold nanoparticles, silica, liposomes, dendrimers, and metal-based agents. We also highlight the mechanism of targeting the tumor cells, and the limitations of different nanotheranostic agents in the field of research for cancer treatment. Due to the complexity in this area, multifunctional and hybrid nanoparticles functionalized with targeted moieties or anti-cancer drugs show the best feature for theranostics that enables them to work on carrying and delivering active materials to the desired area of the requirement for early detection and diagnosis. Non-invasive imaging techniques have a specificity of receptor binding and internalization processes of the nanosystems within the cancer cells. Nanotheranostics may provide the appropriate medicine at the appropriate dose to the appropriate patient at the appropriate time.
Collapse
Affiliation(s)
- Deepshikha Gupta
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India.
| | - Priyanka Roy
- Department of Chemistry, Jamia Hamdard University, New Delhi, 110062, India
| | - Rishabh Sharma
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India
| | - Richa Kasana
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India
| | - Pragati Rathore
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India
| | - Tejendra Kumar Gupta
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India
| |
Collapse
|
170
|
Wang Z, Li W, Li F, Xiao R. An update of predictive biomarkers related to WEE1 inhibition in cancer therapy. J Cancer Res Clin Oncol 2024; 150:13. [PMID: 38231277 PMCID: PMC10794259 DOI: 10.1007/s00432-023-05527-y] [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: 10/09/2023] [Accepted: 11/10/2023] [Indexed: 01/18/2024]
Abstract
PURPOSE WEE1 is a crucial kinase involved in the regulation of G2/M checkpoint within the cell cycle. This article aims to comprehensively review the existing knowledge on the implication of WEE1 as a therapeutic target in tumor progression and drug resistance. Furthermore, we summarize the current predictive biomarkers employed to treat cancer with WEE1 inhibitors. METHODS A systematic review of the literature was conducted to analyze the association between WEE1 inhibition and cancer progression, including tumor advancement and drug resistance. Special attention was paid to the identification and utilization of predictive biomarkers related to therapeutic response to WEE1 inhibitors. RESULTS The review highlights the intricate involvement of WEE1 in tumor progression and drug resistance. It synthesizes the current knowledge on predictive biomarkers employed in WEE1 inhibitor treatments, offering insights into their prognostic significance. Notably, the article elucidates the potential for precision medicine by understanding these biomarkers in the context of tumor treatment outcomes. CONCLUSION WEE1 plays a pivotal role in tumor progression and is a promising therapeutic target. Distinguishing patients that would benefit from WEE1 inhibition will be a major direction of future research.
Collapse
Affiliation(s)
- Zizhuo Wang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wenting Li
- Department of Gynecology, First Affiliated Hospital, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China
| | - Fuxia Li
- Department of Gynecology, First Affiliated Hospital, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China
| | - Rourou Xiao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China.
| |
Collapse
|
171
|
Peng YL, Wang ZY, Zhong RW, Mei SQ, Liu JQ, Tang LB, Guo Z, Ren ZR, Wu L, Deng Y, Chen ZH, Zhou Q, Xu CR. Association of COVID-19 and Lung Cancer: Short-Term and Long-Term Interactions. Cancers (Basel) 2024; 16:304. [PMID: 38254793 PMCID: PMC10813989 DOI: 10.3390/cancers16020304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Background: COVID-19 has been ravaging the globe for more than three years. Due to systemic immunosuppression of anti-tumor therapy, application of chemotherapy and adverse effects of surgery, the short- and long-term prognosis of cancer patients to COVID-19 are of significant concern. Method: This research included three parts of data. The first part of the data came from the public database that covered Veneto residents. The second part of the data included participants in Guangzhou. The third part of the data was used for MR analysis. We assessed the associations by logistic, linear or Cox regression when appropriate. Result: Lung cancer patients with COVID-19 had shorter progression-free survival (PFS) after COVID-19 (Model II: HR: 3.28, 95% CI: 1.6~6.72; Model III: HR: 3.39, 95% CI: 1.45~7.95), compared with lung cancer patients without COVID-19. Targeted therapy patients recovered from SARS-CoV-2 infection more quickly (Model I: β: -0.58, 95% CI: -0.75~-0.41; Model II: β: -0.59, 95% CI: -0.76~-0.41; Model III: β: -0.57; 95% CI: -0.75~-0.40). Conclusions: PFS in lung cancer patients is shortened by COVID-19. The outcome of COVID-19 in lung cancer patients was not significantly different from that of the healthy population. In lung cancer patients, targeted therapy patients had a better outcome of COVID-19, while chemotherapy patients had the worst.
Collapse
Affiliation(s)
- Ying-Long Peng
- School of Medicine, South China University of Technology, Guangzhou 510006, China (R.-W.Z.)
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Zi-Yan Wang
- The First Clinical School, Guangzhou Medical University, Guangzhou 510120, China
| | - Ri-Wei Zhong
- School of Medicine, South China University of Technology, Guangzhou 510006, China (R.-W.Z.)
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Shi-Qi Mei
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Jia-Qi Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Li-Bo Tang
- School of Medicine, South China University of Technology, Guangzhou 510006, China (R.-W.Z.)
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Zhi Guo
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Zi-Rui Ren
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Lv Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Yu Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Qing Zhou
- School of Medicine, South China University of Technology, Guangzhou 510006, China (R.-W.Z.)
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| | - Chong-Rui Xu
- School of Medicine, South China University of Technology, Guangzhou 510006, China (R.-W.Z.)
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510180, China (J.-Q.L.); (Z.G.)
| |
Collapse
|
172
|
Coulter DW, Chhonker YS, Kumar D, Kesherwani V, Aldhafiri WN, McIntyre EM, Alexander G, Ray S, Joshi SS, Li R, Murry DJ, Chaturvedi NK. Marinopyrrole derivative MP1 as a novel anti-cancer agent in group 3 MYC-amplified Medulloblastoma. J Exp Clin Cancer Res 2024; 43:18. [PMID: 38200580 PMCID: PMC10782703 DOI: 10.1186/s13046-024-02944-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: 09/14/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Medulloblastoma (MB) patients with MYC oncogene amplification or overexpression exhibit extremely poor prognoses and therapy resistance. However, MYC itself has been one of the most challenging targets for cancer treatment. Here, we identify a novel marinopyrrole natural derivative, MP1, that shows desirable anti-MYC and anti-cancer activities in MB. METHODS In this study, using MYC-amplified (Group 3) and non-MYC amplified MB cell lines in vitro and in vivo, we evaluated anti-cancer efficacies and molecular mechanism(s) of MP1. RESULTS MP1 significantly suppressed MB cell growth and sphere counts and induced G2 cell cycle arrest and apoptosis in a MYC-dependent manner. Mechanistically, MP1 strongly downregulated the expression of MYC protein. Our results with RNA-seq revealed that MP1 significantly modulated global gene expression and inhibited MYC-associated transcriptional targets including translation/mTOR targets. In addition, MP1 inhibited MYC-target metabolism, leading to declined energy levels. The combination of MP1 with an FDA-approved mTOR inhibitor temsirolimus synergistically inhibited MB cell growth/survival by downregulating the expression of MYC and mTOR signaling components. Our results further showed that as single agents, both MP1 and temsirolimus, were able to significantly inhibit tumor growth and MYC expression in subcutaneously or orthotopically MYC-amplified MB bearing mice. In combination, there were further anti-MB effects on the tumor growth and MYC expression in mice. CONCLUSION These preclinical findings highlight the promise of marinopyrrole MP1 as a novel MYC inhibition approach for MYC-amplified MB.
Collapse
Affiliation(s)
- Don W Coulter
- Department of Pediatrics, Hematology/Oncology Division, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yashpal S Chhonker
- Department of Pharmacy Practice & Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Devendra Kumar
- Department of Pediatrics, Hematology/Oncology Division, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Varun Kesherwani
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wafaa N Aldhafiri
- Department of Pharmacy Practice & Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Erin M McIntyre
- Department of Pediatrics, Hematology/Oncology Division, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Gracey Alexander
- Department of Pediatrics, Hematology/Oncology Division, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sutapa Ray
- Department of Pediatrics, Hematology/Oncology Division, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shantaram S Joshi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rongshi Li
- Department of Pharmacy Practice & Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Daryl J Murry
- Department of Pharmacy Practice & Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nagendra K Chaturvedi
- Department of Pediatrics, Hematology/Oncology Division, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Pediatrics, Hematology and Oncology Division, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
173
|
Laiolo J, Graikioti DG, Barbieri CL, Joray MB, Antoniou AI, Vera DMA, Athanassopoulos CM, Carpinella MC. Novel betulin derivatives as multidrug reversal agents targeting P-glycoprotein. Sci Rep 2024; 14:70. [PMID: 38167542 PMCID: PMC10762177 DOI: 10.1038/s41598-023-49939-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Chemotherapy is a powerful means of cancer treatment but its efficacy is compromised by the emergence of multidrug resistance (MDR), mainly linked to the efflux transporter ABCB1/P-glycoprotein (P-gp). Based on the chemical structure of betulin, identified in our previous work as an effective modulator of the P-gp function, a series of analogs were designed, synthesized and evaluated as a source of novel inhibitors. Compounds 6g and 6i inhibited rhodamine 123 efflux in the P-gp overexpressed leukemia cells, K562/Dox, at concentrations of 0.19 µM and 0.39 µM, respectively, and increased the intracellular accumulation of doxorubicin at the submicromolar concentration of 0.098 µM. Compounds 6g and 6i were able to restore the sensitivity of K562/Dox to Dox at 0.024 µM and 0.19 µM, respectively. Structure-activity relationship analysis and molecular modeling revealed important information about the structural features conferring activity. All the active compounds fitted in a specific region involving mainly transmembrane helices (TMH) 4-6 from one homologous half and TMH 7 and 12 from the other, also showing close contacts with TMH 6 and 12. Compounds that bound preferentially to another region were inactive, regardless of their free energy of binding. It should be noted that compounds 6g and 6i were devoid of toxic effects against peripheral blood mononuclear normal cells and erythrocytes. The data obtained indicates that both compounds might be proposed as scaffolds for obtaining promising P-gp inhibitors for overcoming MDR.
Collapse
Affiliation(s)
- Jerónimo Laiolo
- Fine Chemical and Natural Products Laboratory, IRNASUS CONICET-UCC, Universidad Católica de Córdoba, Córdoba, Argentina
| | - Dafni G Graikioti
- Synthetic Organic Chemistry Laboratory, Department of Chemistry, University of Patras, 26504, Patras, Greece
| | - Cecilia L Barbieri
- Department of Chemistry and Biochemistry, College of Exact and Natural Sciences, Universidad Nacional de Mar del Plata - QUIAMM - INBIOTEC CONICET, Mar del Plata, Argentina
| | - Mariana B Joray
- Fine Chemical and Natural Products Laboratory, IRNASUS CONICET-UCC and CIDIE CONICET-UCC, Universidad Católica de Córdoba, Córdoba, Argentina
| | - Antonia I Antoniou
- Synthetic Organic Chemistry Laboratory, Department of Chemistry, University of Patras, 26504, Patras, Greece
| | - D Mariano A Vera
- Department of Chemistry and Biochemistry, College of Exact and Natural Sciences, Universidad Nacional de Mar del Plata - QUIAMM - INBIOTEC CONICET, Mar del Plata, Argentina.
| | | | - María C Carpinella
- Fine Chemical and Natural Products Laboratory, IRNASUS CONICET-UCC and CIDIE CONICET-UCC, Universidad Católica de Córdoba, Córdoba, Argentina.
| |
Collapse
|
174
|
Ruan D, Wu H, Meng Q, Xu R. Development of antibody-drug conjugates in cancer: Overview and prospects. Cancer Commun (Lond) 2024; 44:3-22. [PMID: 38159059 PMCID: PMC10794012 DOI: 10.1002/cac2.12517] [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: 11/13/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024] Open
Abstract
In recent years, remarkable breakthroughs have been reported on antibody-drug conjugates (ADCs), with 15 ADCs successfully entering the market over the past decade. This substantial development has positioned ADCs as one of the fastest-growing domains in the realm of anticancer drugs, demonstrating their efficacy in treating a wide array of malignancies. Nonetheless, there is still an unmet clinical need for wider application, better efficacy, and fewer side effects of ADCs. An ADC generally comprises an antibody, a linker and a payload, and the combination has profound effects on drug structure, pharmacokinetic profile and efficacy. Hence, optimization of the key components provides an opportunity to develop ADCs with higher potency and fewer side effects. In this review, we comprehensively reviewed the current development and the prospects of ADC, provided an analysis of marketed ADCs and the ongoing pipelines globally as well as in China, highlighted several ADC platforms and technologies specific to different pharmaceutical enterprises and biotech companies, and also discussed the new related technologies, possibility of next-generation ADCs and the directions of clinical research.
Collapse
Affiliation(s)
- Dan‐Yun Ruan
- Department of Clinical ResearchSun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Hao‐Xiang Wu
- Department of Clinical ResearchSun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Qi Meng
- Department of Clinical ResearchSun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Rui‐Hua Xu
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| |
Collapse
|
175
|
Cai F, Xu H, Song S, Wang G, Zhang Y, Qian J, Xu L. Knockdown of Ubiquitin-Conjugating Enzyme E2 T Abolishes the Progression of Head and Neck Squamous Cell Carcinoma by Inhibiting NF-Κb Signaling and inducing Ferroptosis. Curr Protein Pept Sci 2024; 25:577-585. [PMID: 38584528 DOI: 10.2174/0113892037287640240322084946] [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: 11/22/2023] [Revised: 02/17/2024] [Accepted: 02/23/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Ubiquitin-conjugating enzyme 2T (UBE2T) has been reported to be associated with uncontrolled cell growth and tumorigenesis in multiple cancer types. However, the understanding of its regulatory role in the carcinogenesis of Head And Neck Squamous Cell Carcinoma (HNSC) is limited. METHODS UBE2T expression in HNSC patient samples and the correlation between its expression and patients' survival rates were evaluated using The Cancer Genome Atlas (TCGA) database. Cell survival and proliferation were investigated in UM-SCC1 and UM-SCC15 cells infected with control and shUBE2T lentivirus. The xenograft mouse model was established using UM-SCC15 cells to examine HNSC tumorigenesis with or without UBE2T. Western blot, qRT-PCR, and ferroptosis assays were carried out to disclose the interaction between UBE2T and NF-κB signaling and ferroptosis. RESULTS The increased expression of UBE2T was noted in tumor tissues of patients with HNSC, correlating with a significantly reduced overall survival time in this patient cohort. Knockdown of UBE2T inhibited HNSC tumorigenesis and tumor growth. Mechanistically, inhibition of UBE2T suppressed NF-κB signaling and induced ferroptosis in HNSC. CONCLUSION Our study underscores the multifaceted role of UBE2T in HNSC, illuminating its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Feng Cai
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Hongbo Xu
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Shilong Song
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Gengming Wang
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Yajun Zhang
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Jing Qian
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Lu Xu
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| |
Collapse
|
176
|
Misra R, Sivaranjani A, Saleem S, Dash BR. Copper Nanoclusters as Novel Podium for Cancer Detection, Imaging, and Therapy Applications. Crit Rev Ther Drug Carrier Syst 2024; 41:51-80. [PMID: 37938190 DOI: 10.1615/critrevtherdrugcarriersyst.2023044994] [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: 11/09/2023]
Abstract
Nanoclusters (NCs) are meticulously small, kinetically stable, crystalline materials which hold immense potential as multifaceted catalysts for a broad range of biomedical applications. Metal NCs are atomically precise and exist within the range of Fermi wavelength of electrons. They are highly advantageous as functional materials as their physicochemical properties can be customized to meet specific requirements. Copper NCs (CuNCs) are emerging as an efficient substitute to the other existing metal NCs. The synthesis of CuNCs is highly methodical, fast, cost effective and does not involve any complicated manipulation. On the contrary to gold and silver NCs, copper is a vital trace element for humans that can be excreted easily out the body. Further, the relatively inexpensiveness and easy availability of copper aids in potential nanotechnological applications in large quantity. As such, CuNCs have attracted great interest among the research community recently. The modern developments in the strategy, synthesis, surface modifications, and use of CuNCs in diagnosis of disease, imaging and treatment have been discussed in the present review. Approaches to regulate and augment the emission of CuNCs, challenges and drawbacks have also been considered. This review brings to light the multifarious applications of CuNCs and their potential as emerging theranostic agents. It is anticipated that the visions and directions for translating existing developments in CuNCs from the laboratory to the clinic can be further improved and enhanced.
Collapse
Affiliation(s)
- Ranjita Misra
- Department of Biotechnology, School of Sciences, Jain University, Bangalore, Karnataka, India
| | - A Sivaranjani
- Advanced Institute for Wildlife Conservation, Chennai, Tamil Nadu, India
| | - Suraiya Saleem
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai 600036, Tamil Nadu, India
| | - Bignya Rani Dash
- Department of Chemistry, Indian Institute of Technology, Madras, Chennai 600036, Tamil Nadu, India
| |
Collapse
|
177
|
Chen Z, Ding C, Chen J, Zheng S, Li Q. Pan-cancer analysis revealing the multidimensional expression and prognostic and immunologic roles of TGFB1 in cancer. J Int Med Res 2024; 52:3000605231221361. [PMID: 38263929 PMCID: PMC10807329 DOI: 10.1177/03000605231221361] [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: 08/17/2023] [Accepted: 11/16/2023] [Indexed: 01/25/2024] Open
Abstract
OBJECTIVE This study aimed to perform an integrated pan-cancer analysis to characterize the expression patterns, prognostic value, genetic alterations, and immunologic roles of transforming growth factor beta 1 (TGFB1) across diverse human cancer types. METHODS Bioinformatics analyses were conducted using multiple public databases including The Cancer Genome Atlas, Genotype-Tissue Expression, Clinical Proteomic Tumor Analysis Consortium, TIMER2, GEPIA2, cBioPortal, StringDB, and others. Differential expression, survival, immune correlation, and protein interaction network analyses were performed. RESULTS TGFB1 was overexpressed in several tumor types compared with that in normal tissues. High TGFB1 expression was associated with an advanced stage and poorer prognosis in certain cancers. TGFB1 mutations occurred in 1.3% of 10,967 cases surveyed. TGFB1 expression correlated with tumor-infiltrating immune cells and immunotherapy-related genes. CONCLUSIONS This comprehensive multi-omics analysis revealed the complex expression and prognostic landscape of TGFB1 across cancers. TGFB1 is emerging as a potential immunotherapeutic target in certain contexts. Further research should elucidate its multifaceted tumor-promoting and tumor-suppressive mechanisms. Our pan-cancer analysis provides new insights into TGFB1 as a prognostic biomarker and immunotherapeutic target in human cancers, and our findings may guide future preclinical and clinical investigations of TGFB1-directed therapies.
Collapse
Affiliation(s)
- Zhitao Chen
- Department of Hepatobiliary Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Chenchen Ding
- Department of Hepatobiliary Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Jun Chen
- Department of Hepatobiliary Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- School of Medicine, Zhejiang Shuren University, Hangzhou, China
| | - Shusen Zheng
- Department of Hepatobiliary Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Qiyong Li
- Department of Hepatobiliary Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| |
Collapse
|
178
|
Singh A, Mishra R, Mazumder A. Breast cancer and its therapeutic targets: A comprehensive review. Chem Biol Drug Des 2024; 103:e14384. [PMID: 37919259 DOI: 10.1111/cbdd.14384] [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: 05/20/2023] [Revised: 09/14/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Breast cancer is a common and deadly disease, so there is a constant need for research to find efficient targets and therapeutic approaches. Breast cancer can be classified on a molecular and histological base. Breast cancer can be divided into ER (estrogen receptor)-positive and ER-negative, HER2 (human epidermal growth factor receptor2)-positive and HER2-negative subtypes based on the presence of specific biomarkers. Targeting hormone receptors, such as the HER2, progesterone receptor (PR), and ER, is very significant and plays a vital role in the onset and progression of breast cancer. Endocrine treatments and HER2-targeted drugs are examples of targeted therapies now being used against these receptors. Emerging immune-based medicines with promising outcomes in the treatment of breast cancer include immune checkpoint inhibitors, cancer vaccines, and adoptive T-cell therapy. It is also explored how immune cells and the tumor microenvironment affect breast cancer development and treatment response. The major biochemical pathways, signaling cascades, and DNA repair mechanisms that are involved in the development and progression of breast cancer, include the PI3K/AKT/mTOR system, the MAPK pathway, and others. These pathways are intended to be inhibited by a variety of targeted drugs, which are then delivered with the goal of restoring normal cellular function. This review aims to shed light on types of breast cancer with the summarization of different therapeutic approaches which can target different pathways for tailored medicines and better patient outcomes.
Collapse
Affiliation(s)
- Ayushi Singh
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Rakhi Mishra
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| |
Collapse
|
179
|
Jiang Y, Cai Y, Bao Y, Kong X, Jin H. Overexpression of NOP58 Facilitates Proliferation, Migration, Invasion, and Stemness of Non-small Cell Lung Cancer by Stabilizing hsa_circ_0001550. Anticancer Agents Med Chem 2024; 24:1197-1206. [PMID: 38994624 DOI: 10.2174/0118715206293943240615105417] [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: 11/20/2023] [Revised: 05/13/2024] [Accepted: 05/29/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND NOP58 ribonucleoprotein (NOP58) is associated with the recurrence of lung adenocarcinoma. AIMS Few investigations concentrate on the role of NOP58 in non-small cell lung cancer (NSCLC), which is the focus of our current study. METHODS Following transfection, the proliferation, migration, and invasion of NSCLC cells were assessed by 5- ethynyl-2'-deoxyuridine (EdU), wound healing, and transwell assays. The percentage of CD9+ cells was evaluated by flow cytometry assay. Based on target genes and binding sites predicted through bioinformatics analysis, a dual-luciferase reporter assay was performed to verify the targeting relationship between hsa_circ_0001550 and NOP58. The effect of NOP58 overexpression on hsa_circ_0001550 stability was gauged using Actinomycin D. The hsa_circ_0001550 and NOP58 expression levels, as well as protein expressions of CD44, CD133, OCT4, and SOX2 in NSCLC cells were determined by quantitative real-time PCR and Western blot, respectively. RESULTS Hsa_circ_0001550 was remarkably up-regulated in NSCLC cell lines A549 and PC9, silencing of which weakened cell abilities to proliferate, migrate and invade, decreased CD9+ cell ratio, and diminished protein expressions of CD44, CD133, OCT4, and SOX2. NOP58 could bind to hsa_circ_0001550 and stabilize its expression, and NOP58 overexpression partially abrogated hsa_circ_0001550 knockdown-inhibited NSCLC cell proliferation, migration, invasion and stemness. CONCLUSION Overexpression of NOP58 facilitates proliferation, migration, invasion, and stemness of NSCLC cells by stabilizing hsa_circ_0001550, hinting that NOP58 is a novel molecular target for NSCLC therapy.
Collapse
Affiliation(s)
- Yiqian Jiang
- Department of Radiotherapy, Xiaoshan District Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Ying Cai
- Department of Respiratory Medicine, Xiaoshan District Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Yanhong Bao
- Department of Radiotherapy, Xiaoshan District Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Xiangyang Kong
- Department of Radiotherapy, Xiaoshan District Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Haigang Jin
- Department of General Surgery, Xiaoshan District Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
180
|
Xiao T, Wu A, Wang X, Guo Z, Huang F, Cheng X, Shen X, Tao L. Anti-hypertensive and composition as well as pharmacokinetics and tissues distribution of active ingredients from Alpinia zerumbet. Fitoterapia 2024; 172:105753. [PMID: 37992780 DOI: 10.1016/j.fitote.2023.105753] [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: 08/16/2023] [Revised: 10/11/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
Alpinia zerumbet is a food flavor additive and a traditional medicine herb around the world. Several studies have reported that A. zerumbet has excellent effects on a variety of cardiovascular diseases, but its potential hypertensive applications, and pharmacokinetic features of main active substances have not been fully investigated. The mechanism of anti-hypertension with ethyl acetate extracts of A. zerumbet fruits (AZEAE) was evaluated by L-NNA-induced hypertensive rats and L-NAME-injured human umbilical vein endothelial cells (HUVECs). Blood pressure, echocardiographic cardiac index and H&E staining were used to preliminary evaluate the antihypertensive effect of AZEAE, the levels of TNF-α, IL-6, and IL-1β were evaluated by ELISA, and the proteins expression of IL-1β, IL-18, AGTR1, VCAM, iNOS, EDN1 and eNOS were also evaluated. In addition, isolation, identification, and activity screening of bioactive compounds were carried ou. Next, pharmacokinetics and tissues distribution of dihydro-5,6-dehydrokavain (DDK) in vivo were measured, and preliminary absorption mechanism was conducted with Caco-2 cell monolayers. AZEAE remarkably enhanced the state of hypertensive rats. Twelve compounds were isolated and identified, and five compounds were isolated from this plant for the first time. The isolated compounds also exhibited good resistance against injury of HUVECs. Moreover, pharmacokinetics and Caco-2 cell monolayers demonstrated AZEAE had better absorption capacity than DDK, and DDK exhibited differences in tissues distribution and gender difference. This study was the first to assess the potential hypertensive applications of A. zerumbet in vivo and vitro, and the first direct and concise study of the in vivo behavior of DDK and AZEAE.
Collapse
Affiliation(s)
- Ting Xiao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China.
| | - Ai Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Xiaowei Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Zhenghong Guo
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Feilong Huang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Xingyan Cheng
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China.
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China.
| |
Collapse
|
181
|
Yuan Y, Wang W, Luo J, Tang C, Zheng Y, Yu J, Xu H, Zhu M, Hang T, Wang H, Diao X. Metabolite characterisation of the peptide-drug conjugate LN005 in liver S9s by UHPLC-Orbitrap-HRMS. Xenobiotica 2024; 54:1-9. [PMID: 38044881 DOI: 10.1080/00498254.2023.2289635] [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: 09/19/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
LN005 is a peptide-drug conjugate (PDC) targeting glucose-regulated protein 78 (GRP78) to treat several types of cancer, such as breast, colon, and prostate cancer.As a new drug modality, understanding its metabolism and elimination pathways will help us to have a whole picture of it. Currently, there are no metabolic studies on LN005; therefore, this study aimed to investigate the metabolism of LN005, clarify its metabolic profile in the liver S9s of different species, and identify the major metabolic pathways and differences between species.The incubation samples were measured by ultra-high performance liquid chromatography combined with orbitrap tandem mass spectrometry (UHPLC-Orbitrap-HRMS).The results showed that LN005 was metabolised by liver S9s, and four metabolites were identified. The main metabolic pathway of LN005 in liver S9s was oxidative deamination to ketone or hydrolysis. Similar metabolic profiles were observed in mouse, rat, dog, monkey, and human liver S9s, indicating no differences between these four animal species and humans.This study provides information for the structural modification and optimisation of LN005 and affords a reference for subsequent animal experiments and human metabolism of other PDCs.
Collapse
Affiliation(s)
- Yali Yuan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Weiqiang Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Jing Luo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Department of Preparation, Shanghai Whittlong Pharmaceutical Institute Co., Ltd, Shanghai, China
| | - Chongzhuang Tang
- Department of Biotransformation, XenoFinder Co., Ltd, Suzhou, China
| | - Yuandong Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jinghua Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Honghong Xu
- Department of Preparation, Shanghai Whittlong Pharmaceutical Institute Co., Ltd, Shanghai, China
| | - Mingshe Zhu
- Department of Biotransformation, XenoFinder Co., Ltd, Suzhou, China
| | - Taijun Hang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Hao Wang
- National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Xingxing Diao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
182
|
Jiang S, Ma W, Ma C, Zhang Z, Zhang W, Zhang J. An emerging strategy: probiotics enhance the effectiveness of tumor immunotherapy via mediating the gut microbiome. Gut Microbes 2024; 16:2341717. [PMID: 38717360 PMCID: PMC11085971 DOI: 10.1080/19490976.2024.2341717] [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: 01/29/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
The occurrence and progression of tumors are often accompanied by disruptions in the gut microbiota. Inversely, the impact of the gut microbiota on the initiation and progression of cancer is becoming increasingly evident, influencing the tumor microenvironment (TME) for both local and distant tumors. Moreover, it is even suggested to play a significant role in the process of tumor immunotherapy, contributing to high specificity in therapeutic outcomes and long-term effectiveness across various cancer types. Probiotics, with their generally positive influence on the gut microbiota, may serve as effective agents in synergizing cancer immunotherapy. They play a crucial role in activating the immune system to inhibit tumor growth. In summary, this comprehensive review aims to provide valuable insights into the dynamic interactions between probiotics, gut microbiota, and cancer. Furthermore, we highlight recent advances and mechanisms in using probiotics to improve the effectiveness of cancer immunotherapy. By understanding these complex relationships, we may unlock innovative approaches for cancer diagnosis and treatment while optimizing the effects of immunotherapy.
Collapse
Affiliation(s)
- Shuaiming Jiang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wenyao Ma
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Chenchen Ma
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, Shenzhen, PR China
| | - Zeng Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wanli Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| |
Collapse
|
183
|
Sherif AY, Harisa GI, Alanazi FK. The Chimera of TPGS and Nanoscale Lipid Carriers as Lymphatic Drug Delivery Vehicles to Fight Metastatic Cancers. Curr Drug Deliv 2024; 21:525-543. [PMID: 37183467 DOI: 10.2174/1567201820666230512122825] [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: 10/07/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 05/16/2023]
Abstract
The lymphatic system (LS) plays a crucial role in fluid balance, transportation of macromolecules, and immune response. Moreover, LS is a channel for microbial invasion and cancer metastasis. Particularly, solid tumors, including lung, breast, melanoma, and prostate cancers, are metastasized across highways of LS. Subsequently, the fabrication of chimeric lymphatic drug delivery systems (LDDS) is a promising strategy to fight cancer metastasis and control microbial pandemics. In this regard, LDDS, in terms of PEG-nanoscaled lipid carriers, elicited a revolution during the COVID-19 pandemic as cargoes for mRNA vaccines. The drug delivered by the lymphatic pathway escapes first-pass metabolism and enhances the drug's bioavailability. Ample approaches, including synthesis of prodrugs, trigging of chylomicron biosynthesis, and fabrication of nanocarriers, facilitate lymphatic drug delivery. Specifically, nanoscales lipid cargoes have the propensity to lymphatic trafficking. Interestingly, TPGSengineered nanoscale lipid cargoes enhance lymphatic trafficking, increase tissue permeation, and, specifically, uptake. Moreover, they overcome biological barriers, control biodistribution, and enhance organelles localization. Most anticancer agents are non-specific, have low bioavailability, and induced drug resistance. Therefore, TPGS-engineered nanoscale lipid chimeras improve the therapeutic impact of anticancer agents. This review highlights lymphatic cancer metastasis, nanoscales lipid cargoes as LDDS, and their influence on lymphatic trafficking, besides the methods of LDD studies.
Collapse
Affiliation(s)
- Abdelrahman Y Sherif
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Biochemistry and Molecular Biology, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
184
|
Liu L, Xue W. Thalidomide suppresses migration and invasion of colorectal cancer cells by inhibiting HOXB7-mediated activation of the Wnt/β-catenin signaling pathway. Chem Biol Drug Des 2024; 103:e14434. [PMID: 38230780 DOI: 10.1111/cbdd.14434] [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: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
Heaps of studies have verified the effects of thalidomide (THA) on colorectal cancer (CRC). Howbeit, the corresponding mechanism awaits illustration, which is the foothold of this study. Following the treatment of 0, 1.94, 7.75, or 19.36 μM THA, CRC cell viability, apoptosis, migration, and invasion were evaluated by methyl tetrazolium, flow cytometry, wound-healing, and transwell assays. Homeobox B7 (HOXB7) expression in CRC was analyzed and detected by bioinformatics analysis, quantitative real-time PCR or western blot. After the corresponding transfection or treatment with inhibitor of catenin-responsive transcription-3 (iCRT-3), abovementioned CRC cell biological behaviors as well as expression levels of HOXB7 and β-catenin were evaluated. 7.75 and 19.36 μM THA dwindled CRC cell viability, migration, and invasion, and facilitated apoptosis. HOXB7 upregulation was detected in CRC cells, which promoted the viability, migration, invasion, and β-catenin expression, and weakened the apoptosis of CRC cells. Also, HOXB7 upregulation counteracted the effects of THA on CRC cells. iCRT-3 restrained β-catenin expression, viability, migration, and invasion, whereas promoting the apoptosis of CRC cells. In addition, iCRT-3 antagonized the effects of overexpressed HOXB7 on CRC cells. THA inhibits the migration and invasion of CRC cells, which is achieved by suppressing HOXB7-mediated activation of Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Liyang Liu
- Department of Anoretal, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wusong Xue
- Department of Anoretal, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
185
|
Sabale P, Waghmare S, Potey L, Khedekar P, Sabale V, Rarokar N, Chikhale R, Palekar R. Novel targeting strategies on signaling pathways of colorectal cancer. COLORECTAL CANCER 2024:489-531. [DOI: 10.1016/b978-0-443-13870-6.00017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
186
|
Zheng X, Song X, Zhu G, Pan D, Li H, Hu J, Xiao K, Gong Q, Gu Z, Luo K, Li W. Nanomedicine Combats Drug Resistance in Lung Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308977. [PMID: 37968865 DOI: 10.1002/adma.202308977] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/03/2023] [Indexed: 11/17/2023]
Abstract
Lung cancer is the second most prevalent cancer and the leading cause of cancer-related death worldwide. Surgery, chemotherapy, molecular targeted therapy, immunotherapy, and radiotherapy are currently available as treatment methods. However, drug resistance is a significant factor in the failure of lung cancer treatments. Novel therapeutics have been exploited to address complicated resistance mechanisms of lung cancer and the advancement of nanomedicine is extremely promising in terms of overcoming drug resistance. Nanomedicine equipped with multifunctional and tunable physiochemical properties in alignment with tumor genetic profiles can achieve precise, safe, and effective treatment while minimizing or eradicating drug resistance in cancer. Here, this work reviews the discovered resistance mechanisms for lung cancer chemotherapy, molecular targeted therapy, immunotherapy, and radiotherapy, and outlines novel strategies for the development of nanomedicine against drug resistance. This work focuses on engineering design, customized delivery, current challenges, and clinical translation of nanomedicine in the application of resistant lung cancer.
Collapse
Affiliation(s)
- Xiuli Zheng
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Xiaohai Song
- Department of General Surgery, Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Guonian Zhu
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Haonan Li
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Jiankun Hu
- Department of General Surgery, Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Kai Xiao
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Zhongwei Gu
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Weimin Li
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
187
|
Chung YY, Cheng SJ, Ko HH, Shie WY, Elizabeth Chou HY. Evaluation of the prognostic and therapeutic potential of inhibin beta B for oral squamous cell carcinoma. J Dent Sci 2024; 19:448-454. [PMID: 38303818 PMCID: PMC10829639 DOI: 10.1016/j.jds.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/09/2023] [Indexed: 02/03/2024] Open
Abstract
Background/purpose Oral squamous cell carcinoma (OSCC) is a common cancer worldwide, and its metastasis is difficult to predict and prevent. Inhibin beta B (INHBB) protein has been linked to cancer prognosis and epithelial-mesenchymal transition (EMT). However, previous study about INHBB expression focused on patients in a single region while the risk factors vary among regions. This study aimed to provide a broader perspective on INHBB expression in OSCC. Materials and methods Tissue micro-arrays comprising 118 specimens were subjected to immunohistochemistry, and all slides were quantified using StrataQuest software. Results The ratio of INHBB-positive cells to total cells was significantly higher in OSCC samples than in normal samples, and the intensity of INHBB expression was significantly greater in the late-stage OSCC. After classifying specimens into high and low INHBB expression groups, a significant association with clinical staging was found. Though a previous study suggested that menin regulates INHBB, menin expression was not detected in specimens. Conclusion The ratio of INHBB-positive cells in OSCC may be druggable for targeting tumor cells or assisting in diagnosis, and the intensity of INHBB expression may provide prognostic information for predicting potential metastasis. Moreover, the regulatory mechanism of INHBB in OSCC remains unclear and requires further investigation.
Collapse
Affiliation(s)
- Yao-Yu Chung
- Graduate Institute of Oral Biology, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Jung Cheng
- Graduate Institute of Oral Biology, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Hui-Hsin Ko
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital Hsin-Chu Branch, College of Medicine, National Taiwan University, Hsin-Chu, Taiwan
| | - Wan-Yi Shie
- Graduate Institute of Oral Biology, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Yi Elizabeth Chou
- Graduate Institute of Oral Biology, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
188
|
Li K, Ji YM, Guo JL, Guo Q. Biological functions and molecular mechanisms of LINC00961 in human cancer. ALL LIFE 2023. [DOI: 10.1080/26895293.2023.2174707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Affiliation(s)
- Kai Li
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital of Jianyang, Jianyang, Sichuan, People’s Republic of China
| | - Yan-Mei Ji
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Jia-Long Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Qiang Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| |
Collapse
|
189
|
Liu Y, Yin Z, Wang Y, Chen H. Exploration and validation of key genes associated with early lymph node metastasis in thyroid carcinoma using weighted gene co-expression network analysis and machine learning. Front Endocrinol (Lausanne) 2023; 14:1247709. [PMID: 38144565 PMCID: PMC10739373 DOI: 10.3389/fendo.2023.1247709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023] Open
Abstract
Background Thyroid carcinoma (THCA), the most common endocrine neoplasm, typically exhibits an indolent behavior. However, in some instances, lymph node metastasis (LNM) may occur in the early stages, with the underlying mechanisms not yet fully understood. Materials and methods LNM potential was defined as the tumor's capability to metastasize to lymph nodes at an early stage, even when the tumor volume is small. We performed differential expression analysis using the 'Limma' R package and conducted enrichment analyses using the Metascape tool. Co-expression networks were established using the 'WGCNA' R package, with the soft threshold power determined by the 'pickSoftThreshold' algorithm. For unsupervised clustering, we utilized the 'ConsensusCluster Plus' R package. To determine the topological features and degree centralities of each node (protein) within the Protein-Protein Interaction (PPI) network, we used the CytoNCA plugin integrated with the Cytoscape tool. Immune cell infiltration was assessed using the Immune Cell Abundance Identifier (ImmuCellAI) database. We applied the Least Absolute Shrinkage and Selection Operator (LASSO), Support Vector Machine (SVM), and Random Forest (RF) algorithms individually, with the 'glmnet,' 'e1071,' and 'randomForest' R packages, respectively. Ridge regression was performed using the 'oncoPredict' algorithm, and all the predictions were based on data from the Genomics of Drug Sensitivity in Cancer (GDSC) database. To ascertain the protein expression levels and subcellular localization of genes, we consulted the Human Protein Atlas (HPA) database. Molecular docking was carried out using the mcule 1-click Docking server online. Experimental validation of gene and protein expression levels was conducted through Real-Time Quantitative PCR (RT-qPCR) and immunohistochemistry (IHC) assays. Results Through WGCNA and PPI network analysis, we identified twelve hub genes as the most relevant to LNM potential from these two modules. These 12 hub genes displayed differential expression in THCA and exhibited significant correlations with the downregulation of neutrophil infiltration, as well as the upregulation of dendritic cell and macrophage infiltration, along with activation of the EMT pathway in THCA. We propose a novel molecular classification approach and provide an online web-based nomogram for evaluating the LNM potential of THCA (http://www.empowerstats.net/pmodel/?m=17617_LNM). Machine learning algorithms have identified ERBB3 as the most critical gene associated with LNM potential in THCA. ERBB3 exhibits high expression in patients with THCA who have experienced LNM or have advanced-stage disease. The differential methylation levels partially explain this differential expression of ERBB3. ROC analysis has identified ERBB3 as a diagnostic marker for THCA (AUC=0.89), THCA with high LNM potential (AUC=0.75), and lymph nodes with tumor metastasis (AUC=0.86). We have presented a comprehensive review of endocrine disruptor chemical (EDC) exposures, environmental toxins, and pharmacological agents that may potentially impact LNM potential. Molecular docking revealed a docking score of -10.1 kcal/mol for Lapatinib and ERBB3, indicating a strong binding affinity. Conclusion In conclusion, our study, utilizing bioinformatics analysis techniques, identified gene modules and hub genes influencing LNM potential in THCA patients. ERBB3 was identified as a key gene with therapeutic implications. We have also developed a novel molecular classification approach and a user-friendly web-based nomogram tool for assessing LNM potential. These findings pave the way for investigations into the mechanisms underlying differences in LNM potential and provide guidance for personalized clinical treatment plans.
Collapse
Affiliation(s)
- Yanyan Liu
- Department of General Surgery, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, China
| | - Zhenglang Yin
- Department of General Surgery, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, China
| | - Yao Wang
- Digestive Endoscopy Department, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haohao Chen
- Department of General Surgery, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, China
| |
Collapse
|
190
|
Sakshi S, Dey S, Chowdhury S, Ray S. Characterization of a Zeolite-Y-Encapsulated Zn(II)Salmphen Complex with Targeted Anticancer Property. ACS APPLIED MATERIALS & INTERFACES 2023; 15:55518-55532. [PMID: 38010148 DOI: 10.1021/acsami.3c13955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Resistance and severe side effects of classical chemotherapeutic drugs are major challenges to cancer therapy. New therapeutic agents and combination therapy are considered potential solutions that enhance the efficacy of the drug as well as reduce drug resistance. The success of a platinum-based anticancer drug, cisplatin, has paved the way to explore metal-centered anticancer therapeutic agents. Herein, the zeolite-Y-encapsulated Zn(II)Salmphen complex is synthesized using a flexible ligand approach. The Zn(II)Salmphen complex and its encapsulation within the supercage of zeolite-Y were characterized by elemental analysis, Fourier transform infrared (FTIR) spectroscopy, UV-vis, fluorescence, powder X-ray diffraction (PXRD), scanning electron microscopy (SEM), X-ray photoelectron spectroscopy (XPS), NMR, and high-resolution mass spectrometry (HRMS) techniques. Elemental analysis, PXRD, and SEM, all together confirm the integrity of the zeolite framework after the encapsulation of Zn(II)Salmphen complex in it, and elemental analysis provides the Si/Al ratio and Zn content present. FTIR and XPS studies indicate the successful encapsulation of the complex. NMR and HRMS studies confirm that the Zn(II)Salmphen complex is dimer; however, within the supercage of zeolite-Y, it is expected to exist as a monomer. The extent of structural modification of the encapsulated Zn(II)Salmphen complex is intimated by electronic spectroscopic studies. The free-state Zn(II)Salmphen is a fluorescent complex, and even the encapsulated Zn(II)Salmphen complex, when taken in dimethyl sulfoxide (DMSO), shows fluorescence. In comparison to cisplatin, encapsulated Zn(II)Salmphen complex displays comparable cytotoxicity (IC50 = 2.0 ± 0.5 μg/mL at 48 h) toward breast cancer cell line, whereas free Zn(II)Salmphen has better cytotoxicity (IC50 = 1.5 ± 0.5 μg/mL at 48 h). Importantly, elemental analysis has revealed that the IC50 value, if calculated only in terms of Zn(II)Salmphen within Zn(II)Salmphen-Y, is as low as 54.59 ng/mL, indicating a very high efficacy of the drug. Interestingly, a 48 h treatment with the encapsulated Zn(II)Salmphen complex shows no toxicity toward immortal noncancerous keratinocyte cells (HaCaT), whereas cisplatin has an IC50 value of 1.75 ± 0.5 μg/mL. Internalization studies indicate that zeolite-Y targets cancer cells better than it does noncancerous ones. Hence, cellular uptake of the zeolite-encapsulated Zn(II)Salmphen complex in cancer cells is more than that in HaCaT cells, resulting in the generation of more reactive oxygen species and cell death. Significant upregulation of DNA damage response protein indicates that DNA-damage-induced cellular apoptosis could be the mechanism of drug action. Overall, the zeolite-encapsulated Zn(II)Salmphen complex could be a better alternative to the traditional drug cisplatin with minimal effect on noncancerous HaCaT cells and can also be utilized as a fluorescent probe in exploring the mechanistic pathway of its activity against cancer cells.
Collapse
|
191
|
Hawas SS, El-Sayed SM, Elzahhar PA, Moustafa MA. New 2-alkoxycyanopyridine derivatives as inhibitors of EGFR, HER2, and DHFR: Synthesis, anticancer evaluation, and molecular modeling studies. Bioorg Chem 2023; 141:106874. [PMID: 37769524 DOI: 10.1016/j.bioorg.2023.106874] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/07/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
New series of substituted 2-alkoxycyanopyridine derivatives were synthesized and evaluated for their in vitro and in vivo anticancer activities. Comparing the evaluated activities against cancer cell lines to the broad-spectrum anticancer doxorubicin, and the kinase inhibitor sorafenib, compounds 3a, 4b, 4c, 7a, and 8d demonstrated superior anticancer efficacy with elevated safety profiles and selectivity indices, particularly against MCF7 breast cancer. For exploration of their mechanism of action, assays for inhibition of EGFR, HER2 kinase, and DHFR were performed. The promising synthesized compounds exhibited potent dual kinase EGFR/HER2 inhibitory activity with IC50values of 0.248/0.156 μM for 4b and 0.138/0.092 μM for 4c. Additionally, with IC50 values of 0.138 and 0.193 M, respectively, 4b and 4c had the greatest DHFR inhibitory activity that was comparable to methotrexate. In the MCF7 breast cancer cell line, they caused arrest at the S phase of the cell cycle and exhibited apoptosis induction activity. With restored caspase-3 immunoexpression, the anti-breast cancer assay performed in vivo of 4b and 4c demonstrated a substantial decrease in tumor volume. Results from molecular modeling were in agreement with biological assays proving the importance of the 3-caynopyridine, two substituted phenyl rings attached to central pyridine ring, and propoxy side chain moieties for binding with the receptors. As 4c works by inhibiting both EGFR/HER2 kinase, DHFR enzymes, in addition to cellular apoptosis, it could be viewed as a model of compounds possessing a multi-targeting anticancer activity. Collectively, compounds 4b and 4c might represent prototypes for further development as anticancer molecules.
Collapse
Affiliation(s)
- Samia S Hawas
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Selwan M El-Sayed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Perihan A Elzahhar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mohamed A Moustafa
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
192
|
Lei S, He J, Gao P, Wang Y, Hui H, An Y, Tian J. Magnetic Particle Imaging-Guided Hyperthermia for Precise Treatment of Cancer: Review, Challenges, and Prospects. Mol Imaging Biol 2023; 25:1020-1033. [PMID: 37789103 DOI: 10.1007/s11307-023-01856-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 10/05/2023]
Abstract
Magnetic particle imaging (MPI) is a novel quantitative imaging technique using the nonlinear magnetization behavior of magnetic nanoparticles (MNPs) to determine their local concentration. Magnetic fluid hyperthermia (MFH) is a promising non-invasive therapy using the heating effects of MNPs. MPI-MFH is expected to enable real-time MPI guidance, localized MFH, and non-invasive temperature monitoring, which shows great potential for precise treatment of cancer. In this review, we introduce the fundamentals of MPI and MFH and their applications in the treatment of cancer. Also, we discuss the challenges and prospects of MPI-MFH.
Collapse
Affiliation(s)
- Siao Lei
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
| | - Jie He
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
| | - Pengli Gao
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
| | - Yueqi Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
| | - Yu An
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China.
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China.
| | - Jie Tian
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China.
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China.
- Zhuhai Precision Medical Center, Zhuhai People's Hospital, Affiliated With Jinan University, Zhuhai, 519000, China.
| |
Collapse
|
193
|
Mannan A, Kakkar C, Dhiman S, Singh TG. Advancing the frontiers of adaptive cell therapy: A transformative mechanistic journey from preclinical to clinical settings. Int Immunopharmacol 2023; 125:111095. [PMID: 37875038 DOI: 10.1016/j.intimp.2023.111095] [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: 08/24/2023] [Revised: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023]
Abstract
Although the concept of using the patient's immune system to combat cancer has been around for a while, it is only in recent times that substantial progress has been achieved in this field. Over the last ten years, there has been a significant advancement in the treatment of cancer through immune checkpoint blockade. This treatment has been approved for multiple types of tumors. Another approach to modifying the immune system to detect tumor cells and fight them off is adaptive cell therapy (ACT). This therapy involves using T cells that have been modified with either T cell receptors (TCR) or chimeric antigen receptors (CAR) to target the tumor cells. ACT has demonstrated encouraging outcomes in different types of tumors, and clinical trials are currently underway worldwide to enhance this form of treatment. This review focuses on the advancements that have been made in ACT from preclinical to clinical settings till now.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Chirag Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
194
|
Yang L, Atakhanova N, Arellano MTC, Mohamed MY, Hani T, Fahdil AA, Castillo-Acobo RY, Juyal A, Hussein AK, Amin AH, Pecho RDC, Akhavan-Sigari R. Translational research of new developments in targeted therapy of colorectal cancer. Pathol Res Pract 2023; 252:154888. [PMID: 37948996 DOI: 10.1016/j.prp.2023.154888] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
A severe global health concern is the rising incidence and mortality rate of colorectal cancer (CRC). Chemotherapy, which is typically used to treat CRC, is known to have limited specificity and can have noticeable side effects. A paradigm shift in cancer treatment has been brought about by the development of targeted therapies, which has led to the appearance of pharmacological agents with improved efficacy and decreased toxicity. Epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), and BRAF are among the molecular targets covered in this review that are used in targeted therapy for CRC. The current discussion also covers advancements in targeted therapeutic approaches, such as antibody-drug conjugates, immune checkpoint inhibitors, and chimeric antigen receptor (CAR) T-cell therapy. A review of the clinical trials and application of these particular therapies in treating CRC is also done. Despite the improvements in targeted therapy for CRC, problems such as drug resistance and patient selection remain to be solved. Despite this, targeted therapies have offered fresh possibilities for identifying and treating CRC, paving the way for the development of personalized medicine and extending the life expectancy and general well-being of CRC patients.
Collapse
Affiliation(s)
- Lei Yang
- Department of Clinical Laboratory, People's Hospital of Chongqing Liangjiang New Area, Chongqing 401121, China
| | - Nigora Atakhanova
- Head of the Department of Oncology, Tashkent Medical Academy, Tashkent 100109, Uzbekistan
| | | | | | - Thamer Hani
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | - Ali A Fahdil
- Medical technical college, Al-Farahidi University, Iraq
| | | | - Ashima Juyal
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| | | | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland
| |
Collapse
|
195
|
Calvillo-Rodríguez KM, Lorenzo-Anota HY, Rodríguez-Padilla C, Martínez-Torres AC, Scott-Algara D. Immunotherapies inducing immunogenic cell death in cancer: insight of the innate immune system. Front Immunol 2023; 14:1294434. [PMID: 38077402 PMCID: PMC10701401 DOI: 10.3389/fimmu.2023.1294434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
Cancer immunotherapies include monoclonal antibodies, cytokines, oncolytic viruses, cellular therapies, and other biological and synthetic immunomodulators. These are traditionally studied for their effect on the immune system's role in eliminating cancer cells. However, some of these therapies have the unique ability to directly induce cytotoxicity in cancer cells by inducing immunogenic cell death (ICD). Unlike general immune stimulation, ICD triggers specific therapy-induced cell death pathways, based on the release of damage-associated molecular patterns (DAMPs) from dying tumour cells. These activate innate pattern recognition receptors (PRRs) and subsequent adaptive immune responses, offering the promise of sustained anticancer drug efficacy and durable antitumour immune memory. Exploring how onco-immunotherapies can trigger ICD, enhances our understanding of their mechanisms and potential for combination strategies. This review explores the complexities of these immunotherapeutic approaches that induce ICD, highlighting their implications for the innate immune system, addressing challenges in cancer treatment, and emphasising the pivotal role of ICD in contemporary cancer research.
Collapse
Affiliation(s)
- Kenny Misael Calvillo-Rodríguez
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Helen Yarimet Lorenzo-Anota
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
- The Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, NL, Mexico
| | - Cristina Rodríguez-Padilla
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Ana Carolina Martínez-Torres
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Daniel Scott-Algara
- Département d'Immunologie, Unité de Biologie Cellulaire des Lymphocytes, Pasteur Institute, Paris, France
| |
Collapse
|
196
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
197
|
Barnieh FM, Galuska SP, Loadman PM, Ward S, Falconer RA, El-Khamisy SF. Cancer-specific glycosylation of CD13 impacts its detection and activity in preclinical cancer tissues. iScience 2023; 26:108219. [PMID: 37942010 PMCID: PMC10628746 DOI: 10.1016/j.isci.2023.108219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/14/2023] [Accepted: 10/12/2023] [Indexed: 11/10/2023] Open
Abstract
Harnessing the differences between cancer and non-cancer tissues presents new opportunities for selective targeting by anti-cancer drugs. CD13, a heavily glycosylated protein, is one example with significant unmet clinical potential in cancer drug discovery. Despite its high expression and activity in cancers, CD13 is also expressed in many normal tissues. Here, we report differential tissue glycosylation of CD13 across tissues and demonstrate for the first time that the nature and pattern of glycosylation of CD13 in preclinical cancer tissues are distinct compared to normal tissues. We identify cancer-specific O-glycosylation of CD13, which selectively blocks its detection in cancer models but not in normal tissues. In addition, the metabolism activity of cancer-expressed CD13 was observed to be critically dependent on its unique glycosylation. Thus, our data demonstrate the existence of discrete cancer-specific CD13 glycoforms and propose cancer-specific CD13 glycoforms as a clinically useful target for effective cancer-targeted therapy.
Collapse
Affiliation(s)
- Francis M. Barnieh
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Sebastian P. Galuska
- Institute for Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, Dummerstorf, Germany
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | | | - Robert A. Falconer
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Sherif F. El-Khamisy
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
198
|
Jabbari A, Sameiyan E, Yaghoobi E, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM. Aptamer-based targeted delivery systems for cancer treatment using DNA origami and DNA nanostructures. Int J Pharm 2023; 646:123448. [PMID: 37757957 DOI: 10.1016/j.ijpharm.2023.123448] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/14/2023] [Accepted: 09/24/2023] [Indexed: 09/29/2023]
Abstract
Due to the limitations of conventional cancer treatment methods, nanomedicine has appeared as a promising alternative, allowing improved drug targeting and decreased drug toxicity. In the development of cancer nanomedicines, among various nanoparticles (NPs), DNA nanostructures are more attractive because of their precisely controllable size, shape, excellent biocompatibility, programmability, biodegradability, and facile functionalization. Aptamers are introduced as single-stranded RNA or DNA molecules with recognize their corresponding targets. So, incorporating aptamers into DNA nanostructures led to influential vehicles for bioimaging and biosensing as well as targeted cancer therapy. In this review, the recent developments in the application of aptamer-based DNA origami and DNA nanostructures in advanced cancer treatment have been highlighted. Some of the main methods of cancer treatment are classified as chemo-, gene-, photodynamic- and combined therapy. Finally, the opportunities and problems for targeted DNA aptamer-based nanocarriers for medicinal applications have also been discussed.
Collapse
Affiliation(s)
- Atena Jabbari
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Sameiyan
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Yaghoobi
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie-Curie, Ottawa, ON K1N 6N5, Canada
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
199
|
Chiang CW, Lin YS, Chang FL, Lin TY, Tsai KC, HuangFu WC, Lee YC. Single-chain fragment antibody disrupting the EphA4 function as a therapeutic drug for gastric cancer. Biochem Biophys Res Commun 2023; 680:161-170. [PMID: 37741263 DOI: 10.1016/j.bbrc.2023.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023]
Abstract
Studies have shown that the high expression of EphA4 in gastric cancer tissues may correlate with unfavorable clinical pathological characteristics. Therefore, EphA4 may be an effective target for treating gastric cancer in addition to HER-2/neu. In this study, generated scFv S3 can bind endogenous EphA4 of gastric cancer cells and has significant membrane staining. Additionally, scFv S3 binding to EphA4 inhibits the growth and migration of cancer cells and the growth induction that ephrinA1 generates in gastric cancer cells. We found that EphA4 molecules may degrade through antibody treatment of cells, and the increase in LAMP1 and LAMP2 indicates that lysosome is involved in the degradation. The scFv S3 administration leads to the signals pAKT, pERK, and pSTAT3 decrease in cancer cells. The xenograft model of HER-2/neu low expressing gastric cancer cell SNU-16 exhibits better therapeutic effects by scFv S3 than trastuzumab scFv. The scFv S3 administration in vivo can degrade EphA4 molecules in tumor tissues, decreasing Ki67 and increasing cleaved C3 molecule expression. Furthermore, we identified and validated that scFv S3 generates essential ionic bonding with R162 on EphA4. The antibody may provide effective treatment for patients with gastric cancer and abnormal activation or overexpression of EphA4 signaling.
Collapse
Affiliation(s)
- Chen-Wei Chiang
- Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yun-Shih Lin
- Department of Psychiatry, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Fu-Ling Chang
- Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tsai-Yu Lin
- Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chun HuangFu
- Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Yu-Ching Lee
- Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
200
|
Beljkas M, Ilic A, Cebzan A, Radovic B, Djokovic N, Ruzic D, Nikolic K, Oljacic S. Targeting Histone Deacetylases 6 in Dual-Target Therapy of Cancer. Pharmaceutics 2023; 15:2581. [PMID: 38004560 PMCID: PMC10674519 DOI: 10.3390/pharmaceutics15112581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Histone deacetylases (HDACs) are the major regulators of the balance of acetylation of histone and non-histone proteins. In contrast to other HDAC isoforms, HDAC6 is mainly involved in maintaining the acetylation balance of many non-histone proteins. Therefore, the overexpression of HDAC6 is associated with tumorigenesis, invasion, migration, survival, apoptosis and growth of various malignancies. As a result, HDAC6 is considered a promising target for cancer treatment. However, none of selective HDAC6 inhibitors are in clinical use, mainly because of the low efficacy and high concentrations used to show anticancer properties, which may lead to off-target effects. Therefore, HDAC6 inhibitors with dual-target capabilities represent a new trend in cancer treatment, aiming to overcome the above problems. In this review, we summarize the advances in tumor treatment with dual-target HDAC6 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Katarina Nikolic
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (A.I.); (A.C.); (B.R.); (N.D.); (D.R.)
| | - Slavica Oljacic
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (A.I.); (A.C.); (B.R.); (N.D.); (D.R.)
| |
Collapse
|