1
|
Kahraman T, Yagci FC, Ceylan A, Calik A, Tarman IO, Kiran F. A novel trivalent inactivated Salmonella vaccine formulated with CpG ODNs to enhance the cellular immunity in chickens. Poult Sci 2025; 104:105024. [PMID: 40120243 PMCID: PMC11981755 DOI: 10.1016/j.psj.2025.105024] [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: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
This study aimed to develop and evaluate a CpG oligodeoxynucleotide (CpG ODN)-adjuvanted trivalent inactivated Salmonella vaccine including S. enterica subsp. Enterica serovar Typhimurium, Salmonella enterica subsp. Enterica serovar Enteritidis, and Salmonella enterica serotype Infantis, for its immunogenic efficacy in chickens. The immunomodulatory effects of various CpG ODNs were assessed based on proinflammatory cytokine secretion and the expression levels of CD80, CD86, and MHC-II in the chicken cell lines HD11 and DT40. According to the results, CpG ODNs D35 3CG PO, D35 3CG MB, 1466 Acore PO, 1466 Acore MB, and K3 which exhibited non-cytotoxicity in both HD11 and DT40 cell lines, were selected for vaccine formulation. To evaluate their effects under in vivo conditions, chicks (n = 25) were randomly assigned to fourteen groups (G1: only sterile pyrogen-free saline solution, G2: only inactivated vaccine, G3: inactivated vaccine with 150 mg/dose of ALUM, G4: commercial Salenvac T vaccine, G5-G14: various experimental vaccine formulations which included different CpG ODNs combined with inactivated bacterial strains, with or without ALUM). Immune responses were analyzed through serological assays for antigen-specific antibody titers and ex vivo splenocyte cultures for cytokine secretion. Flow cytometry was performed to assess T-cell activation and IFN-γ production. The results demonstrated that the CpG ODNs-adjuvanted vaccine formulations significantly enhanced both humoral and cellular immunity compared to the commercial vaccine. Specifically, the Vac#5+ ALUM formulation, which included the K3 CpG ODN, induced robust antibody responses against Salmonella antigens and significantly increased IFN-γ secretion, nearly two-fold higher than the commercial vaccine. This effect was primarily mediated by CD4+ helper and CD8+ cytotoxic T cells. These findings highlight the potential of CpG ODNs as effective vaccine adjuvants in poultry. To the best of our knowledge, this is the first study to investigate the use of CpG ODNs as adjuvants in inactivated Salmonella vaccine formulations. Future studies should focus on evaluating the long-term protective efficacy of this vaccine formulation and its ability to provide cross-protection against a broader spectrum of Salmonella serovars.
Collapse
Affiliation(s)
- Tamer Kahraman
- THORVACS Biotechnology, Bilkent Cyberpark, 06800, Ankara, Turkey.
| | - Fuat Cem Yagci
- ARBO Biotechnology, SL6 8BY, Maidenhead, United Kingdom.
| | - Ahmet Ceylan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, 06110, Ankara, Turkey.
| | - Ali Calik
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Ankara University, 06110, Ankara, Turkey.
| | | | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, 06100, Ankara, Turkey.
| |
Collapse
|
2
|
Shariati A, Khezrpour A, Shariati F, Afkhami H, Yarahmadi A, Alavimanesh S, Kamrani S, Modarressi MH, Khani P. DNA vaccines as promising immuno-therapeutics against cancer: a new insight. Front Immunol 2025; 15:1498431. [PMID: 39872522 PMCID: PMC11769820 DOI: 10.3389/fimmu.2024.1498431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Cancer is one of the leading causes of mortality around the world and most of our conventional treatments are not efficient enough to combat this deadly disease. Harnessing the power of the immune system to target cancer cells is one of the most appealing methods for cancer therapy. Nucleotide-based cancer vaccines, especially deoxyribonucleic acid (DNA) cancer vaccines are viable novel cancer treatments that have recently garnered significant attention. DNA cancer vaccines are made of plasmid molecules that encode tumor-associated or tumor-specific antigens (TAAs or TSAs), and possibly some other immunomodulatory adjuvants such as pro-inflammatory interleukins. Following the internalization of plasmids into cells, their genes are expressed and the tumor antigens are loaded on major histocompatibility molecules to be presented to T-cells. After the T-cells have been activated, they will look for tumor antigens and destroy the tumor cells upon encountering them. As with any other treatment, there are pros and cons associated with using these vaccines. They are relatively safe, usually well-tolerated, stable, easily mass-produced, cost-effective, and easily stored and transported. They can induce a systemic immune response effective on both the primary tumor and metastases. The main disadvantage of DNA vaccines is their poor immunogenicity. Several approaches including structural modification, combination therapy with conventional and novel cancer treatments (such as chemotherapy, radiotherapy, and immune checkpoint blockade (ICB)), and the incorporation of adjuvants into the plasmid structure have been studied to enhance the vaccine's immunogenicity and improve the clinical outcome of cancer patients. In this review, we will discuss some of the most promising optimization strategies and examine some of the important trials regarding these vaccines.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
3
|
Giudice GC, Sonpavde GP. Vaccine approaches to treat urothelial cancer. Hum Vaccin Immunother 2024; 20:2379086. [PMID: 39043175 PMCID: PMC11268260 DOI: 10.1080/21645515.2024.2379086] [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: 03/28/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024] Open
Abstract
Bladder cancer (BC) accounts for about 4% of all malignancies. Non-muscle-invasive BC, 75% of cases, is treated with transurethral resection and adjuvant intravesical instillation, while muscle-invasive BC warrants cisplatin-based perioperative chemotherapy. Although immune-checkpoint inhibitors, antibody drug conjugates and targeted agents have provided dramatic advances, metastatic BC remains a generally incurable disease and clinical trials continue to vigorously evaluate novel molecules. Cancer vaccines aim at activating the patient's immune system against tumor cells. Several means of delivering neoantigens have been developed, including peptides, antigen-presenting cells, virus, or nucleic acids. Various improvements are constantly being explored, such as adjuvants use and combination strategies. Nucleic acids-based vaccines are increasingly gaining attention in recent years, with promising results in other malignancies. However, despite the recent advantages, numerous obstacles persist. This review is aimed at describing the different types of cancer vaccines, their evaluations in UC patients and the more recent innovations in this field.
Collapse
Affiliation(s)
- Giulia Claire Giudice
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Guru P. Sonpavde
- AdventHealth Cancer Institute, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
4
|
Agrez M, Chandler C, Thurecht KJ, Fletcher NL, Liu F, Subramaniam G, Howard CB, Parker S, Turner D, Rzepecka J, Knox G, Nika A, Hall AM, Gooding H, Gallagher L. A novel immunomodulating peptide with potential to complement oligodeoxynucleotide-mediated adjuvanticity in vaccination strategies. Sci Rep 2024; 14:26737. [PMID: 39501043 PMCID: PMC11538426 DOI: 10.1038/s41598-024-78150-7] [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: 06/27/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
The identification of adjuvants to improve vaccination efficacy is a major unmet need. One approach is to augment the functionality of dendritic cells (DCs) by using Toll-like receptor-9 (TLR9) agonists such as cytosine-phosphate-guanine oligodeoxynucleotides (CpG ODNs) as adjuvants. Another approach is adjuvant selection based on production of bioactive interleukin-12 (IL-12). We report a D-peptide isomer, designated D-15800, that induces monocyte differentiation to the DC phenotype in vitro and more effectively stimulates IL-12p70 production upon T cell receptor (TCR) activation than the L-isomer. In the absence of TCR activation and either IL-12p70 or interleukin-2 production, only D-15800 activates CD4+ T and natural killer cells. In the presence of CpG ODN, D-15800 synergistically enhances production of interferon-alpha (IFN-α). Taken together with its biostability in human serum and depot retention upon injection, co-delivery of D-15800 with TLR9 agonists could serve to improve vaccine efficacy.
Collapse
Affiliation(s)
- Michael Agrez
- InterK Peptide Therapeutics Limited, Lane Cove West, NSW, Australia.
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia.
| | | | - Kristofer J Thurecht
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Feifei Liu
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Gayathri Subramaniam
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Stephen Parker
- InterK Peptide Therapeutics Limited, Lane Cove West, NSW, Australia
| | | | | | - Gavin Knox
- Concept Life Sciences, Edinburgh, Scotland
| | | | | | | | | |
Collapse
|
5
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [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: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
6
|
Quispe-Salcedo A, Yamazaki T, Ohshima H. Effects of Synthetic Toll-Like Receptor 9 Ligand Molecules on Pulpal Immunomodulatory Response and Repair after Injuries. Biomolecules 2024; 14:931. [PMID: 39199319 PMCID: PMC11353191 DOI: 10.3390/biom14080931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Synthetic oligodeoxynucleotides (ODNs) containing unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG-ODNs) are ligand molecules for Toll-like receptor 9 (TLR9), which is expressed by odontoblasts in vitro and dental pulp cells. This study determined the effects of CpG-ODNs on pulpal immunomodulatory response and repair following injury. Briefly, the upper right first molars of three-week-old mice were extracted, immersed in Type A (D35) or B (K3) CpG-ODN solutions (0.1 or 0.8 mM) for 30 min, and then replanted. Pulpal healing and immunomodulatory activity were assessed by hematoxylin-eosin and AZAN staining, as well as immunohistochemistry. One week following the operation, inflammatory reactions occurred in all of the experimental groups; however, re-revascularization and newly formed hard tissue deposition were observed in the pulp chamber of all groups at week 2. A positive trend in the expression of immune cell markers was observed toward the CpG-ODN groups at 0.1 mM. Our data suggest that synthetic CpG-ODN solutions at low concentrations may evoke a long-lasting macrophage-TLR9-mediated pro-inflammatory, rather than anti-inflammatory, response in the dental pulp to modulate the repair process and hard tissue formation. Further studies are needed to determine the effects of current immunomodulatory agents in vitro and in vivo and develop treatment strategies for dental tissue regeneration.
Collapse
Affiliation(s)
- Angela Quispe-Salcedo
- Division of Anatomy and Cell Biology of the Hard Tissue, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Tomohiko Yamazaki
- Research Center for Macromolecules and Biomaterials, National Institute of Material Sciences (NIMS), Tsukuba 305-0047, Japan;
| | - Hayato Ohshima
- Division of Anatomy and Cell Biology of the Hard Tissue, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| |
Collapse
|
7
|
Mohite P, Yadav V, Pandhare R, Maitra S, Saleh FM, Saleem RM, Al-malky HS, Kumarasamy V, Subramaniyan V, Abdel-Daim MM, Uti DE. Revolutionizing Cancer Treatment: Unleashing the Power of Viral Vaccines, Monoclonal Antibodies, and Proteolysis-Targeting Chimeras in the New Era of Immunotherapy. ACS OMEGA 2024; 9:7277-7295. [PMID: 38405458 PMCID: PMC10882662 DOI: 10.1021/acsomega.3c06501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/27/2024]
Abstract
In the realm of cancer immunotherapy, a profound evolution has ushered in sophisticated strategies that encompass both traditional cancer vaccines and emerging viral vaccines. This comprehensive Review offers an in-depth exploration of the methodologies, clinical applications, success stories, and future prospects of these approaches. Traditional cancer vaccines have undergone significant advancements utilizing diverse modalities such as proteins, peptides, and dendritic cells. More recent innovations have focused on the physiological mechanisms enabling the human body to recognize and combat precancerous and malignant cells, introducing specific markers like peptide-based anticancer vaccines targeting tumor-associated antigens. Moreover, cancer viral vaccines, leveraging engineered viruses to stimulate immune responses against specific antigens, exhibit substantial promise in inducing robust and enduring immunity. Integration with complementary therapeutic methods, including monoclonal antibodies, adjuvants, and radiation therapy, has not only improved survival rates but also deepened our understanding of viral virulence. Recent strides in vaccine design, encompassing oncolytic viruses, virus-like particles, and viral vectors, mark the frontier of innovation. While these advances hold immense potential, critical challenges must be addressed, such as strategies for immune evasion, potential off-target effects, and the optimization of viral genomes. In the landscape of immunotherapy, noteworthy innovations take the spotlight from the use of immunomodulatory agents for the enhancement of innate and adaptive immune collaboration. The emergence of proteolysis-targeting chimeras (PROTACs) as precision tools for cancer therapy is particularly exciting. With a focus on various cancers, from melanoma to formidable solid tumors, this Review critically assesses types of cancer vaccines, mechanisms, barriers in vaccine therapy, vaccine efficacy, safety profiles, and immune-related adverse events, providing a nuanced perspective on the underlying mechanisms involving cytotoxic T cells, natural killer cells, and dendritic cells. The Review also underscores the transformative potential of cutting-edge technologies such as clinical studies, molecular sequencing, and artificial intelligence in advancing the field of cancer vaccines. These tools not only expedite progress but also emphasize the multidimensional and rapidly evolving nature of this research, affirming its profound significance in the broader context of cancer therapy.
Collapse
Affiliation(s)
- Popat Mohite
- AETs
St. John Institute of Pharmacy and Research, Palghar, Maharashtra 401404, India
| | - Vaishnavi Yadav
- AETs
St. John Institute of Pharmacy and Research, Palghar, Maharashtra 401404, India
| | - Ramdas Pandhare
- MESs
College of Pharmacy, Sonai Tal-Newasa, Maharashtra 414105, India
| | - Swastika Maitra
- Center
for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India
- Department
of Microbiology, Adamas University, Kolkata 700 126, West Bengal, India
| | - Fayez M. Saleh
- Department
of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rasha Mohammed Saleem
- Department
of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha 65431, Saudi Arabia
| | - Hamdan S. Al-malky
- Regional
Drug Information Center, Ministry of Health, Jeddah 11176, Saudi Arabia
| | - Vinoth Kumarasamy
- Department
of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology
Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar
Sunway, 47500 Selangor
Darul Ehsan, Malaysia
- Center
for Transdisciplinary Research, Department of Pharmacology, Savetha
Dental College, Savetha Institute of Medical and Technical Sciences, Savetha University, Chennai, Tamil Nadu 600077, India
| | - Mohamed M. Abdel-Daim
- Department
of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box
6231, Jeddah 21442, Saudi Arabia
- Pharmacology
Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Daniel E. Uti
- Department
of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Benue State 970001, Nigeria
| |
Collapse
|
8
|
Goyal F, Chattopadhyay A, Navik U, Jain A, Reddy PH, Bhatti GK, Bhatti JS. Advancing Cancer Immunotherapy: The Potential of mRNA Vaccines As a Promising Therapeutic Approach. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202300255] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Indexed: 01/11/2025]
Abstract
AbstractmRNA vaccines have long been recognized for their ability to induce robust immune responses. The discovery that mRNA vaccines may also contribute to antitumor immunity has made them a promising therapeutic approach against cancer. Recent advances in understanding of immune system are precious in developing therapeutic strategies that target pathways involved in tumor survival and progression, leading to the most reliable therapeutic strategies in cancer treatment history. Among all traditional cancer treatments, cancer immunotherapies are less toxic and more effective, even in advanced or recurrent stages of cancer. Recent advancements in genomics and machine learning algorithms give new insight into vaccine development. mRNA vaccines are designed to interfere with stimulator of interferon genes (STING) and tumor‐infiltrating lymphocytes pathways, activating more CD8+ T‐cells involved in destroying tumor cells and inhibiting tumor growth. A stronger immune response can be achieved by incorporating immunological adjuvants alongside mRNA. Nonformulated or vehicle‐based mRNA vaccines, when combined with adjuvants, efficiently express tumor antigens through antigen‐presenting cells and stimulate both innate and adaptive immune responses. Codelivery with additional immunotherapeutic agents, such as checkpoint inhibitors, further enhances the efficacy of mRNA vaccines. This article focuses on the current clinical approaches and challenges to consider when developing mRNA‐based vaccine technology for cancer treatment.
Collapse
Affiliation(s)
- Falak Goyal
- Laboratory of Translational Medicine and Nanotherapeutics Department of Human Genetics and Molecular Medicine School of Health Sciences Central University of Punjab Bathinda 151401 India
| | - Anandini Chattopadhyay
- Laboratory of Translational Medicine and Nanotherapeutics Department of Human Genetics and Molecular Medicine School of Health Sciences Central University of Punjab Bathinda 151401 India
| | - Umashanker Navik
- Department of Pharmacology School of Health Sciences Central University of Punjab Bathinda 151401 India
| | - Aklank Jain
- Department of Zoology Central University of Punjab Bathinda Punjab 151401 India
| | - P. Hemachandra Reddy
- Department of Internal Medicine Texas Tech University Health Sciences Center Lubbock TX 79430 USA
- Department of Pharmacology and Neuroscience and Garrison Institute on Aging Texas Tech University Health Sciences Center Lubbock TX 79430 USA
- Department of Public Health Graduate School of Biomedical Sciences Texas Tech University Health Sciences Center Lubbock TX 79430 USA
- Department of Neurology Texas Tech University Health Sciences Center Lubbock TX 79430 USA
- Department of Speech Language, and Hearing Sciences Texas Tech University Health Sciences Center Lubbock TX 79430 USA
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology University Institute of Applied Health Sciences Chandigarh University Mohali 140413 India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics Department of Human Genetics and Molecular Medicine School of Health Sciences Central University of Punjab Bathinda 151401 India
| |
Collapse
|
9
|
Bhat AA, Goyal A, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Singh M, Rohilla S, Saini TK, Kukreti N, Meenakshi DU, Fuloria NK, Sekar M, Gupta G. Uncovering the complex role of interferon-gamma in suppressing type 2 immunity to cancer. Cytokine 2023; 171:156376. [PMID: 37748333 DOI: 10.1016/j.cyto.2023.156376] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023]
Abstract
Cancer involves cells' abnormal growth and ability to invade or metastasize to different body parts. Cancerous cells can divide uncontrollably and spread to other areas through the lymphatic or circulatory systems. Tumors form when malignant cells clump together in an uncontrolled manner. In this context, the cytokine interferon-gamma (IFN-γ) is crucial in regulating immunological responses, particularly malignancy. While IFN-γ is well-known for its potent anti-tumor effects by activating type 1 immunity, recent research has revealed its ability to suppress type 2 immunity, associated with allergy and inflammatory responses. This review aims to elucidate the intricate function of IFN-γ in inhibiting type 2 immune responses to cancer. We explore how IFN-γ influences the development and function of immune cells involved in type 2 immunity, such as mast cells, eosinophils, and T-helper 2 (Th2) cells. Additionally, we investigate the impact of IFN-mediated reduction of type 2 immunity on tumor development, metastasis, and the response to immunotherapeutic interventions. To develop successful cancer immunotherapies, it is crucial to comprehend the complex interplay between type 2 and type 1 immune response and the regulatory role of IFN-γ. This understanding holds tremendous promise for the development of innovative treatment approaches that harness the abilities of both immune response types to combat cancer. However, unraveling the intricate interplay between IFN-γ and type 2 immunity in the tumor microenvironment will be essential for achieving this goal.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U. P., India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Mahaveer Singh
- Swami Keshvanand Institute of Pharmacy (SKIP), Raiser, Bikaner, 334022, India
| | - Suman Rohilla
- SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, 122505, India
| | - Tarun Kumar Saini
- Dept. Of Neurosurgery ICU, Lok Nayak Hospital, New Delhi (Govt. Of NCT Of Delhi), New Delhi, India
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | | | | | - Mahendran Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Gaurav Gupta
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
10
|
Lee CW, Bakre A, Olivier TL, Alvarez-Narvaez S, Harrell TL, Conrad SJ. Toll-like Receptor Ligands Enhance Vaccine Efficacy against a Virulent Newcastle Disease Virus Challenge in Chickens. Pathogens 2023; 12:1230. [PMID: 37887747 PMCID: PMC10610171 DOI: 10.3390/pathogens12101230] [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: 09/20/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
To enhance the efficacy of the current Newcastle disease vaccine, we have selected potential adjuvants that target well-characterized pattern recognition receptors: the toll-like receptors (TLRs). Imiquimod is a small-molecule activator of TLR7, which is a sensor of dsDNA. ODN-1826 is a mimetic of CpG DNA and ligates TLR21 (a chicken homologue of TLR9 in mammals). The activation of TLRs leads to antiviral responses, including the induction of type I interferons (IFNs). In this study, birds were vaccinated intranasally with a live LaSota strain with or without imiquimod or ODN-1826 (50 µg/bird). Two weeks after vaccination, the birds were challenged with a virulent Newcastle disease virus (chicken/CA/212676/2002). Both adjuvants (imiquimod or ODN-1826) induced higher and more uniform antibody titers among vaccinated birds compared with the live vaccine-alone group. In addition, adjuvanted vaccines demonstrated greater protective efficacy in terms of the reduction in virus-shedding titer and the number of birds shedding the challenge virus at 2 and 4 days post-challenge. A differential expression of antiviral and immune-related genes was observed among groups from tissues (Harderian gland, trachea, cecal tonsil, and spleen) collected 1 and 3 days after treatment. These results demonstrate the potential of TLR-targeted adjuvants as mucosal vaccine enhancers and warrant a further characterization of immune correlates and optimization for efficacy.
Collapse
Affiliation(s)
- Chang-Won Lee
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, GA 30605, USA; (A.B.); (T.L.O.)
| | - Abhijeet Bakre
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, GA 30605, USA; (A.B.); (T.L.O.)
| | - Timothy L. Olivier
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, GA 30605, USA; (A.B.); (T.L.O.)
| | - Sonsiray Alvarez-Narvaez
- Endemic Poultry Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, GA 30605, USA; (S.A.-N.); (T.L.H.); (S.J.C.)
| | - Telvin L. Harrell
- Endemic Poultry Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, GA 30605, USA; (S.A.-N.); (T.L.H.); (S.J.C.)
| | - Steven J. Conrad
- Endemic Poultry Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, GA 30605, USA; (S.A.-N.); (T.L.H.); (S.J.C.)
| |
Collapse
|
11
|
Kaczmarek M, Poznańska J, Fechner F, Michalska N, Paszkowska S, Napierała A, Mackiewicz A. Cancer Vaccine Therapeutics: Limitations and Effectiveness-A Literature Review. Cells 2023; 12:2159. [PMID: 37681891 PMCID: PMC10486481 DOI: 10.3390/cells12172159] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
In recent years, there has been a surge of interest in tumor microenvironment-associated cancer vaccine therapies. These innovative treatments aim to activate and enhance the body's natural immune response against cancer cells by utilizing specific antigens present in the tumor microenvironment. The goal is to achieve a complete clinical response, where all measurable cancer cells are either eliminated or greatly reduced in size. With their potential to revolutionize cancer treatment, these therapies represent a promising avenue for researchers and clinicians alike. Despite over 100 years of research, the success of therapeutic cancer vaccines has been variable, particularly in advanced cancer patients, with various limitations, including the heterogeneity of the tumor microenvironment, the presence of immunosuppressive cells, and the potential for tumor escape mechanisms. Additionally, the effectiveness of these therapies may be limited by the variability of the patient's immune system response and the difficulty in identifying appropriate antigens for each patient. Despite these challenges, tumor microenvironment-targeted vaccine cancer therapies have shown promising results in preclinical and clinical studies and have the potential to become a valuable addition to current cancer treatment and "curative" options. While chemotherapeutic and monoclonal antibody treatments remain popular, ongoing research is needed to optimize the design and delivery of these therapies and to identify biomarkers that can predict response and guide patient selection. This comprehensive review explores the mechanisms of cancer vaccines, various delivery methods, and the role of adjuvants in improving treatment outcomes. It also discusses the historical background of cancer vaccine research and examines the current state of major cancer vaccination immunotherapies. Furthermore, the limitations and effectiveness of each vaccine type are analyzed, providing insights into the future of cancer vaccine development.
Collapse
Affiliation(s)
- Mariusz Kaczmarek
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, 61-866 Poznań, Poland
| | - Justyna Poznańska
- Scientific Society of Cancer Immunology, Poznań University of Medical Sciences, 61-866 Poznań, Poland; (J.P.)
| | - Filip Fechner
- Scientific Society of Cancer Immunology, Poznań University of Medical Sciences, 61-866 Poznań, Poland; (J.P.)
| | - Natasza Michalska
- Scientific Society of Cancer Immunology, Poznań University of Medical Sciences, 61-866 Poznań, Poland; (J.P.)
| | - Sara Paszkowska
- Scientific Society of Cancer Immunology, Poznań University of Medical Sciences, 61-866 Poznań, Poland; (J.P.)
| | - Adrianna Napierała
- Scientific Society of Cancer Immunology, Poznań University of Medical Sciences, 61-866 Poznań, Poland; (J.P.)
| | - Andrzej Mackiewicz
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, 61-866 Poznań, Poland
| |
Collapse
|
12
|
Guo TJF, Singhera GK, Leung JM, Dorscheid DR. Airway Epithelial-Derived Immune Mediators in COVID-19. Viruses 2023; 15:1655. [PMID: 37631998 PMCID: PMC10458661 DOI: 10.3390/v15081655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The airway epithelium, which lines the conducting airways, is central to the defense of the lungs against inhaled particulate matter and pathogens such as SARS-CoV-2, the virus that causes COVID-19. Recognition of pathogens results in the activation of an innate and intermediate immune response which involves the release of cytokines and chemokines by the airway epithelium. This response can inhibit further viral invasion and influence adaptive immunity. However, severe COVID-19 is characterized by a hyper-inflammatory response which can give rise to clinical presentations including lung injury and lead to acute respiratory distress syndrome, viral pneumonia, coagulopathy, and multi-system organ failure. In response to SARS-CoV-2 infection, the airway epithelium can mount a maladaptive immune response which can delay viral clearance, perpetuate excessive inflammation, and contribute to the pathogenesis of severe COVID-19. In this article, we will review the barrier and immune functions of the airway epithelium, how SARS-CoV-2 can interact with the epithelium, and epithelial-derived cytokines and chemokines and their roles in COVID-19 and as biomarkers. Finally, we will discuss these immune mediators and their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Tony J. F. Guo
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
| | - Gurpreet K. Singhera
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Janice M. Leung
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Delbert R. Dorscheid
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
13
|
Piri-Gharaghie T, Doosti A, Mirzaei SA. Novel adjuvant nano-vaccine induced immune response against Acinetobacter baumannii. AMB Express 2023; 13:31. [PMID: 36905472 PMCID: PMC10008545 DOI: 10.1186/s13568-023-01531-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Developing adjuvant vaccines to combat rising multidrug-resistant (MDR) Acinetobacter baumannii (A. baumannii) infections is a promising and cost-effective approach. The aim of this analysis was to construct a pDNA-CPG C274-adjuvant nano-vaccine and investigate its immunogenicity and protection in BALB/c mice. The CPG ODN C274 adjuvant was chemically synthesized and cloned into pcDNA3.1( +), and the cloning was verified using PCR and BamHI/EcoRV restriction enzyme digestion. Then, utilizing a complex coacervation approach, pDNA-CPG C274 was encapsulated by chitosan (CS) nanoparticles (NPs). TEM and DLS are used to explore the properties of the pDNA/CSNP complex. TLR-9 pathway activation was investigated in human HEK-293 and RAW 264.7 mouse cells. The vaccine's immunogenicity and immune-protective effectiveness were investigated in BALB/c mice. The pDNA-CPG C274/CSNPs were small (mean size 79.21 ± 0.23 nm), positively charged (+ 38.87 mV), and appeared to be spherical. A continuous slow release pattern was achieved. TLR-9 activation was greatest in the mouse model with CpG ODN (C274) at concentrations of 5 and 10 μg/ml with 56% and 55%, respectively (**P < 0.01). However, in HEK-293 human cells, by increasing the concentration of CpG ODN (C274) from 1 to 50 μg/ml, the activation rate of TLR-9 also increased, so that the highest activation rate (81%) was obtained at the concentration of 50 μg/ml (***P < 0.001). pDNA-CPG C274/CSNPs immunized BALB/c mice produced increased amounts of total-IgG, as well as IFN-γ and IL-1B in serum samples, compared to non-encapsulated pDNA-CPG C274. Furthermore, liver and lung injuries, as well as bacterial loads in the liver, lung, and blood, were reduced, and BALB/c mice immunized with pDNA-CPG C274/CSNPs showed potent protection (50-75%) against acute fatal Intraperitoneal A. baumannii challenge. pDNA-CPG C274/CSNPs evoked total-IgG antibodies, Th1 cellular immunity, and the TLR-9 pathway, as well as protection against an acute fatal A. baumannii challenge. Our findings suggest that this nano-vaccine is a promising approach for avoiding A. baumannii infection when used as a powerful adjuvant.
Collapse
Affiliation(s)
- Tohid Piri-Gharaghie
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Seyed Abbas Mirzaei
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.,Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
14
|
Domingues C, Cabral C, Jarak I, Veiga F, Dourado M, Figueiras A. The Debate between the Human Microbiota and Immune System in Treating Aerodigestive and Digestive Tract Cancers: A Review. Vaccines (Basel) 2023; 11:vaccines11030492. [PMID: 36992076 DOI: 10.3390/vaccines11030492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/23/2023] Open
Abstract
The human microbiota comprises a group of microorganisms co-existing in the human body. Unbalanced microbiota homeostasis may impact metabolic and immune system regulation, shrinking the edge between health and disease. Recently, the microbiota has been considered a prominent extrinsic/intrinsic element of cancer development and a promising milestone in the modulation of conventional cancer treatments. Particularly, the oral cavity represents a yin-and-yang target site for microorganisms that can promote human health or contribute to oral cancer development, such as Fusobacterium nucleatum. Moreover, Helicobacter pylori has also been implicated in esophageal and stomach cancers, and decreased butyrate-producing bacteria, such as Lachnospiraceae spp. and Ruminococcaceae, have demonstrated a protective role in the development of colorectal cancer. Interestingly, prebiotics, e.g., polyphenols, probiotics (Faecalibacterium, Bifidobacterium, Lactobacillus, and Burkholderia), postbiotics (inosine, butyrate, and propionate), and innovative nanomedicines can modulate antitumor immunity, circumventing resistance to conventional treatments and could complement existing therapies. Therefore, this manuscript delivers a holistic perspective on the interaction between human microbiota and cancer development and treatment, particularly in aerodigestive and digestive cancers, focusing on applying prebiotics, probiotics, and nanomedicines to overcome some challenges in treating cancer.
Collapse
Affiliation(s)
- Cátia Domingues
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Cristiana Cabral
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ivana Jarak
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Marília Dourado
- Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Health Studies and Research of the University of Coimbra (CEISUC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Studies and Development of Continuous and Palliative Care (CEDCCP), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
15
|
Nihashi Y, Miyoshi M, Umezawa K, Shimosato T, Takaya T. Identification of a Novel Osteogenetic Oligodeoxynucleotide (osteoDN) That Promotes Osteoblast Differentiation in a TLR9-Independent Manner. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1680. [PMID: 35630904 PMCID: PMC9145662 DOI: 10.3390/nano12101680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022]
Abstract
Dysfunction of bone-forming cells, osteoblasts, is one of the causes of osteoporosis. Accumulating evidence has indicated that oligodeoxynucleotides (ODNs) designed from genome sequences have the potential to regulate osteogenic cell fate. Such osteogenetic ODNs (osteoDNs) targeting and activating osteoblasts can be the candidates of nucleic acid drugs for osteoporosis. In this study, the ODN library derived from the Lacticaseibacillus rhamnosus GG genome was screened to determine its osteogenetic effect on murine osteoblast cell line MC3T3-E1. An 18-base ODN, iSN40, was identified to enhance alkaline phosphatase activity of osteoblasts within 48 h. iSN40 also induced the expression of osteogenic genes such as Msx2, osterix, collagen type 1α, osteopontin, and osteocalcin. Eventually, iSN40 facilitated calcium deposition on osteoblasts at the late stage of differentiation. Intriguingly, the CpG motif within iSN40 was not required for its osteogenetic activity, indicating that iSN40 functions in a TLR9-independent manner. These data demonstrate that iSN40 serves as a novel osteogenetic ODN (osteoDN) that promotes osteoblast differentiation. iSN40 provides a potential seed of the nucleic acid drug that activating osteoblasts for osteoporosis therapy.
Collapse
Affiliation(s)
- Yuma Nihashi
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (Y.N.); (T.S.)
| | - Mana Miyoshi
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
| | - Koji Umezawa
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Takeshi Shimosato
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (Y.N.); (T.S.)
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Tomohide Takaya
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (Y.N.); (T.S.)
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| |
Collapse
|
16
|
Emerging Roles of Green-Synthesized Chalcogen and Chalcogenide Nanoparticles in Cancer Theranostics. JOURNAL OF NANOTECHNOLOGY 2022. [DOI: 10.1155/2022/6176610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The last few decades have seen an overwhelming increase in the amount of research carried out on the use of inorganic nanoparticles. More fascinating is the tremendous progress made in the use of chalcogen and chalcogenide nanoparticles in cancer theranostics. These nanomaterials, which were initially synthesized through chemical methods, have now been efficiently produced using different plant materials. The paradigm shift towards the biogenic route of nanoparticle synthesis stems from its superior advantages of biosafety, eco-friendliness, and simplicity, among others. Despite a large number of reviews available on inorganic nanoparticle synthesis through green chemistry, there is currently a dearth of information on the green synthesis of chalcogens and chalcogenides for cancer research. Nanoformulations involving chalcogens such as sulfur, selenium, and tellurium and their respective chalcogenides have recently emerged as promising tools in cancer therapeutics and diagnosis. Similar to other inorganic nanoparticles, chalcogens and chalcogenides have been synthesized using plant extracts and their purified biomolecules. In this review, we provide an up-to-date discussion of the recent progress that has been made in the plant-mediated synthesis of chalcogens and chalcogenides with a special focus on their application in cancer theranostics.
Collapse
|
17
|
Machida K, Tahara SM. Immunotherapy and Microbiota for Targeting of Liver Tumor-Initiating Stem-like Cells. Cancers (Basel) 2022; 14:2381. [PMID: 35625986 PMCID: PMC9139909 DOI: 10.3390/cancers14102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 02/08/2023] Open
Abstract
Cancer contains tumor-initiating stem-like cells (TICs) that are resistant to therapies. Hepatocellular carcinoma (HCC) incidence has increased twice over the past few decades, while the incidence of other cancer types has trended downward globally. Therefore, an understanding of HCC development and therapy resistance mechanisms is needed for this incurable malignancy. This review article describes links between immunotherapies and microbiota in tumor-initiating stem-like cells (TICs), which have stem cell characteristics with self-renewal ability and express pluripotency transcription factors such as NANOG, SOX2, and OCT4. This review discusses (1) how immunotherapies fail and (2) how gut dysbiosis inhibits immunotherapy efficacy. Gut dysbiosis promotes resistance to immunotherapies by breaking gut immune tolerance and activating suppressor immune cells. Unfortunately, this leads to incurable recurrence/metastasis development. Personalized medicine approaches targeting these mechanisms of TIC/metastasis-initiating cells are emerging targets for HCC immunotherapy and microbiota modulation therapy.
Collapse
Affiliation(s)
- Keigo Machida
- Southern California Research Center for ALPD and Cirrhosis, Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 2011 Zonal Ave., 503C-HMR, Los Angeles, CA 90033, USA;
| | | |
Collapse
|
18
|
Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol 2022; 15:28. [PMID: 35303904 PMCID: PMC8931585 DOI: 10.1186/s13045-022-01247-x] [Citation(s) in RCA: 380] [Impact Index Per Article: 126.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/03/2022] [Indexed: 02/08/2023] Open
Abstract
Research on tumor immunotherapy has made tremendous progress in the past decades, with numerous studies entering the clinical evaluation. The cancer vaccine is considered a promising therapeutic strategy in the immunotherapy of solid tumors. Cancer vaccine stimulates anti-tumor immunity with tumor antigens, which could be delivered in the form of whole cells, peptides, nucleic acids, etc. Ideal cancer vaccines could overcome the immune suppression in tumors and induce both humoral immunity and cellular immunity. In this review, we introduced the working mechanism of cancer vaccines and summarized four platforms for cancer vaccine development. We also highlighted the clinical research progress of the cancer vaccines, especially focusing on their clinical application and therapeutic efficacy, which might hopefully facilitate the future design of the cancer vaccine.
Collapse
Affiliation(s)
- Jian Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Minyang Fu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
19
|
Carbajal-Valenzuela IA, Medina-Ramos G, Caicedo-Lopez LH, Jiménez-Hernández A, Ortega-Torres AE, Contreras-Medina LM, Torres-Pacheco I, Guevara-González RG. Extracellular DNA: Insight of a Signal Molecule in Crop Protection. BIOLOGY 2021; 10:biology10101022. [PMID: 34681122 PMCID: PMC8533321 DOI: 10.3390/biology10101022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary Agriculture systems use multiple chemical treatments to prevent pests and diseases, and to fertilize plants and eliminate weeds around the crop. These practices are less accepted by the consumers each day, mostly because of the associated environmental, health, and ecological impact; thus, new sustainable green technologies are being developed to replace the use of chemical products. Among green technologies for agriculture practices, the use of plant elicitors represents an alternative with great potential, and extracellular DNA has shown beneficial effects on important production traits such as defence mechanisms, plant growth and development, and secondary metabolites production that results in yield increment and better-quality food. In this review, we reunite experimental evidence of the natural effect that extracellular DNA has on plants. We also aim to contribute a step closer to the agricultural application of extracellular DNA. Additionally, we suggest that extracellular DNA can have a biostimulant effect on plants, and can be applied as a highly sustainable treatment contributing to the circular economy of primary production. Abstract Agricultural systems face several challenges in terms of meeting everyday-growing quantities and qualities of food requirements. However, the ecological and social trade-offs for increasing agricultural production are high, therefore, more sustainable agricultural practices are desired. Researchers are currently working on diverse sustainable techniques based mostly on natural mechanisms that plants have developed along with their evolution. Here, we discuss the potential agricultural application of extracellular DNA (eDNA), its multiple functioning mechanisms in plant metabolism, the importance of hormetic curves establishment, and as a challenge: the technical limitations of the industrial scale for this technology. We highlight the more viable natural mechanisms in which eDNA affects plant metabolism, acting as a damage/microbe-associated molecular pattern (DAMP, MAMP) or as a general plant biostimulant. Finally, we suggest a whole sustainable system, where DNA is extracted from organic sources by a simple methodology to fulfill the molecular characteristics needed to be applied in crop production systems, allowing the reduction in, or perhaps the total removal of, chemical pesticides, fertilizers, and insecticides application.
Collapse
Affiliation(s)
- Ireri Alejandra Carbajal-Valenzuela
- C. A. Biosystems Engineering, Campus Amazcala, Autonomous University of Queretaro, Carr. Chichimequillas-Amazcala Km 1 S/N, C.P., El Marques, Querétaro 76265, Mexico; (I.A.C.-V.); (L.H.C.-L.); (A.J.-H.); (A.E.O.-T.); (L.M.C.-M.); (I.T.-P.)
| | - Gabriela Medina-Ramos
- Molecular Plant Pathology Laboratory, Polytechnic University of Guanajuato, Cortazar 38496, Mexico
- Correspondence: (G.M.-R.); or (R.G.G.-G.); Tel.: +52-1-461-441-4300 (G.M.-R.); +52-1-442-192-1200 (ext. 6093) (R.G.G.-G.)
| | - Laura Helena Caicedo-Lopez
- C. A. Biosystems Engineering, Campus Amazcala, Autonomous University of Queretaro, Carr. Chichimequillas-Amazcala Km 1 S/N, C.P., El Marques, Querétaro 76265, Mexico; (I.A.C.-V.); (L.H.C.-L.); (A.J.-H.); (A.E.O.-T.); (L.M.C.-M.); (I.T.-P.)
| | - Alejandra Jiménez-Hernández
- C. A. Biosystems Engineering, Campus Amazcala, Autonomous University of Queretaro, Carr. Chichimequillas-Amazcala Km 1 S/N, C.P., El Marques, Querétaro 76265, Mexico; (I.A.C.-V.); (L.H.C.-L.); (A.J.-H.); (A.E.O.-T.); (L.M.C.-M.); (I.T.-P.)
| | - Adrian Esteban Ortega-Torres
- C. A. Biosystems Engineering, Campus Amazcala, Autonomous University of Queretaro, Carr. Chichimequillas-Amazcala Km 1 S/N, C.P., El Marques, Querétaro 76265, Mexico; (I.A.C.-V.); (L.H.C.-L.); (A.J.-H.); (A.E.O.-T.); (L.M.C.-M.); (I.T.-P.)
| | - Luis Miguel Contreras-Medina
- C. A. Biosystems Engineering, Campus Amazcala, Autonomous University of Queretaro, Carr. Chichimequillas-Amazcala Km 1 S/N, C.P., El Marques, Querétaro 76265, Mexico; (I.A.C.-V.); (L.H.C.-L.); (A.J.-H.); (A.E.O.-T.); (L.M.C.-M.); (I.T.-P.)
| | - Irineo Torres-Pacheco
- C. A. Biosystems Engineering, Campus Amazcala, Autonomous University of Queretaro, Carr. Chichimequillas-Amazcala Km 1 S/N, C.P., El Marques, Querétaro 76265, Mexico; (I.A.C.-V.); (L.H.C.-L.); (A.J.-H.); (A.E.O.-T.); (L.M.C.-M.); (I.T.-P.)
| | - Ramón Gerardo Guevara-González
- C. A. Biosystems Engineering, Campus Amazcala, Autonomous University of Queretaro, Carr. Chichimequillas-Amazcala Km 1 S/N, C.P., El Marques, Querétaro 76265, Mexico; (I.A.C.-V.); (L.H.C.-L.); (A.J.-H.); (A.E.O.-T.); (L.M.C.-M.); (I.T.-P.)
- Correspondence: (G.M.-R.); or (R.G.G.-G.); Tel.: +52-1-461-441-4300 (G.M.-R.); +52-1-442-192-1200 (ext. 6093) (R.G.G.-G.)
| |
Collapse
|
20
|
Eusébio D, Neves AR, Costa D, Biswas S, Alves G, Cui Z, Sousa Â. Methods to improve the immunogenicity of plasmid DNA vaccines. Drug Discov Today 2021; 26:2575-2592. [PMID: 34214667 DOI: 10.1016/j.drudis.2021.06.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/31/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
DNA vaccines have emerged as innovative approaches that have great potential to overcome the limitations of current conventional vaccines. Plasmid DNA vaccines are often safer than other vaccines because they carry only antigen genetic information, are more stable and easier to produce, and can stimulate both humoral and cellular immune responses. Although the results of ongoing clinical trials are very promising, some limitations compromise the immunogenicity of these vaccines. Thus, this review describes different strategies that can be explored to improve the immunogenicity of plasmid DNA vaccines, including the optimization of the plasmid vector backbone, the use of different methods for vaccine delivery, the use of alternative administration routes and the inclusion of adjuvants. In combination, these improvements could lead to the successful clinical use of plasmid DNA vaccines.
Collapse
Affiliation(s)
- Dalinda Eusébio
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana R Neves
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Diana Costa
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad 500078, Telangana, India
| | - Gilberto Alves
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX 78712, USA
| | - Ângela Sousa
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
21
|
Overcoming the challenges of tissue delivery for oligonucleotide therapeutics. Trends Pharmacol Sci 2021; 42:588-604. [PMID: 34020790 DOI: 10.1016/j.tips.2021.04.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022]
Abstract
Synthetic therapeutic oligonucleotides (STO) represent the third bonafide platform for drug discovery in the pharmaceutical industry after small molecule and protein therapeutics. So far, thirteen STOs have been approved by regulatory agencies and over one hundred of them are in different stages of clinical trials. STOs hybridize to their target RNA or DNA in cells via Watson-Crick base pairing to exert their pharmacological effects. This unique class of therapeutic agents has the potential to target genes and gene products that are considered undruggable by other therapeutic platforms. However, STOs must overcome several extracellular and intracellular obstacles to interact with their biological RNA targets inside cells. These obstacles include degradation by extracellular nucleases, scavenging by the reticuloendothelial system, filtration by the kidney, traversing the capillary endothelium to access the tissue interstitium, cell-surface receptor-mediated endocytic uptake, and escape from endolysosomal compartments to access the nuclear and/or cytoplasmic compartments where their targets reside. In this review, we present the recent advances in this field with a specific focus on antisense oligonucleotides (ASOs) and siRNA therapeutics.
Collapse
|
22
|
Extracellular DNA in blood products and its potential effects on transfusion. Biosci Rep 2021; 40:222322. [PMID: 32150264 PMCID: PMC7098128 DOI: 10.1042/bsr20192770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/18/2020] [Accepted: 03/03/2020] [Indexed: 02/05/2023] Open
Abstract
Blood transfusions are sometimes necessary after a high loss of blood due to injury or surgery. Some people need regular transfusions due to medical conditions such as haemophilia or cancer. Studies have suggested that extracellular DNA including mitochondrial DNA present in the extracellular milieu of transfused blood products has biological actions that are capable of activating the innate immune systems and potentially contribute to some adverse reactions in transfusion. From the present work, it becomes increasingly clear that extracellular DNA encompassed mitochondrial DNA is far from being biologically inert in blood products. It has been demonstrated to be present in eligible blood products and thus can be transfused to blood recipients. Although the presence of extracellular DNA in human plasma was initially detected in 1948, some aspects have not been fully elucidated. In this review, we summarize the potential origins, clearance mechanisms, relevant structures, and potential role of extracellular DNA in the innate immune responses and its relationship with individual adverse reactions in transfusion.
Collapse
|