1
|
Liu X, Zhang S, Qiu H, Xie ZQ, Tang WF, Chen Y, Wei X. Investigation of high-mobility group box 1 variants with lymph node status and colorectal cancer risk. World J Gastrointest Oncol 2025; 17:102584. [PMID: 40235898 PMCID: PMC11995333 DOI: 10.4251/wjgo.v17.i4.102584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/31/2024] [Accepted: 01/22/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Accumulating studies indicated that maintain nuclei homeostasis was deemed to the protective factors for the occurrence of cancer. Thus, high-mobility group box 1 (HMGB1) might influence the risk and poorer prognoses of colorectal cancer (CRC). AIM This study was designed to investigate whether HMGB1 polymorphisms influence the risk and lymph node metastasis (LNM) of CRC. METHODS Firstly, we designed an investigation with 1003 CRC patients and 1303 cancer-free controls to observe whether HMGB1 rs1412125 T > C and rs1045411 C > T SNPs could influence the risk of cancer. Subsequently, we carried out a correlation-analysis to assess whether these SNPs could alter the risk of LNM. RESULTS The current investigation suggested a relationship of HMGB1 rs1412125 SNP with the increased susceptibility of CRC. In a subgroup analysis, our findings suggested that this SNP could enhance an occurrence of CRC in ≥ 61 years, non-drinker and body mass index < 24 kg/m2 subgroups. However, we found that there was null association between HMGB1 rs1412125 SNP and LNM, even in different CRC region. These observations were confirmed by calculating the power value (more than 0.8). The association of HMGB1 rs1045411 C > T SNP with CRC risk and LNM was not found in any compare. CONCLUSION This study highlights a possible association between HMGB1 rs1412125 polymorphism and the increased risk of CRC. In the future, more studies should be conducted to explore HMGB1 rs1412125 polymorphism in relation to CRC development.
Collapse
Affiliation(s)
- Xin Liu
- Department of General Surgery, Changzhou Third People’s Hospital, Changzhou 213001, Jiangsu Province, China
| | - Sheng Zhang
- Department of General Surgery, Changzhou Third People’s Hospital, Changzhou 213001, Jiangsu Province, China
| | - Hao Qiu
- Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212000, Jiangsu Province, China
| | - Zhi-Qiang Xie
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Wei-Feng Tang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, Jiangsu Province, China
| | - Yu Chen
- Department of Medical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuqing 350014, Fujian Province, China
| | - Xi Wei
- Department of Pathology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu Province, China
| |
Collapse
|
2
|
Malla R, Srilatha M, Muppala V, Farran B, Chauhan VS, Nagaraju GP. Neoantigens and cancer-testis antigens as promising vaccine candidates for triple-negative breast cancer: Delivery strategies and clinical trials. J Control Release 2024; 370:707-720. [PMID: 38744346 DOI: 10.1016/j.jconrel.2024.05.020] [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: 11/30/2023] [Revised: 03/15/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Immunotherapy is gaining prominence as a promising strategy for treating triple-negative breast cancer (TNBC). Neoantigens (neoAgs) and cancer-testis antigens (CTAs) are tumor-specific targets originating from somatic mutations and epigenetic changes in cancer cells. These antigens hold great promise for personalized cancer vaccines, as supported by preclinical and early clinical evidence in TNBC. This review delves into the potential of neoAgs and CTAs as vaccine candidates, emphasizing diverse strategies and delivery approaches. It also highlights the current status of vaccination modalities undergoing clinical trials in TNBC therapy. A comprehensive understanding of neoAgs, CTAs, vaccination strategies, and innovative delivery methods is crucial for optimizing neoAg-based immunotherapies in clinical practice.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati 517502, AP, India
| | - Veda Muppala
- Department of Neuroscience, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Batoul Farran
- Division of Hematology and Oncology, Department of Medicine, Henry Ford Health, Detroit, MI 48202, USA
| | - Virander Singh Chauhan
- Molecular Medicine Group, Molecular Medicines International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
3
|
Keshtpour Amlashi Z, Sheida F, Anbiaee R, Tapak L, Hosseini SR, Mosaedian H, Barati N. The Effect of Melatonin on Increasing the Health Related Quality of Life in Non-Metastatic Breast Cancer Patients: Three-Year Follow up a Clinical Trial. Integr Cancer Ther 2024; 23:15347354241237520. [PMID: 38622853 PMCID: PMC11020700 DOI: 10.1177/15347354241237520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/01/2024] [Accepted: 02/20/2024] [Indexed: 04/17/2024] Open
Abstract
INTRODUCTION Although breast cancer is common worldwide, if diagnosed early and treated on time, the probability of recovery is high and patients often experience a long life. Reducing the quality of life is a common side effect in patients. Melatonin may have an important role in fatigue, sleep disorders and, as a result, the health-related quality of life (HRQoL) in people. About 184 patients with breast cancer were enrolled in 2 groups: intervention with daily melatonin intake of 18 mg for 3 years (93 patients) and the control group with placebo intake (91 patients). Health-related quality of life and the effect of melatonin on increasing that were evaluated with the EORTC QLQ-C30 questionnaire, third edition at the beginning, 2 months later and 3 years after the beginning of the study. RESULTS The general score of the HRQoL was significantly different both in the passage of time and in the comparative study of the 2 groups, and it was better in the melatonin group (P < .05). CONCLUSION Long-term use of 18 mg of melatonin for 3 years in patients with non-metastatic breast cancer can lead to an increase in the patients' quality of life.
Collapse
Affiliation(s)
| | - Fatemeh Sheida
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Robab Anbiaee
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leili Tapak
- Department of Biostatistics, School of Public Health, Modeling of No Communicable Diseases Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | | | - Nastaran Barati
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
4
|
Ghafelehbashi R, Salehi M, Kouhi M, AlizadehNaini A, Sajadi-Javan ZS, Nejatidanesh F. Recent progress in cancer immunotherapy: Application of nano-therapeutic systems. J Drug Deliv Sci Technol 2024; 91:105184. [DOI: 10.1016/j.jddst.2023.105184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Zahedipour F, Jamialahmadi K, Zamani P, Reza Jaafari M. Improving the efficacy of peptide vaccines in cancer immunotherapy. Int Immunopharmacol 2023; 123:110721. [PMID: 37543011 DOI: 10.1016/j.intimp.2023.110721] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Peptide vaccines have shown great potential in cancer immunotherapy by targeting tumor antigens and activating the patient's immune system to mount a specific response against cancer cells. However, the efficacy of peptide vaccines in inducing a sustained immune response and achieving clinical benefit remains a major challenge. In this review, we discuss the current status of peptide vaccines in cancer immunotherapy and strategies to improve their efficacy. We summarize the recent advancements in the development of peptide vaccines in pre-clinical and clinical settings, including the use of novel adjuvants, neoantigens, nano-delivery systems, and combination therapies. We also highlight the importance of personalized cancer vaccines, which consider the unique genetic and immunological profiles of individual patients. We also discuss the strategies to enhance the immunogenicity of peptide vaccines such as multivalent peptides, conjugated peptides, fusion proteins, and self-assembled peptides. Although, peptide vaccines alone are weak immunogens, combining peptide vaccines with other immunotherapeutic approaches and developing novel approaches such as personalized vaccines can be promising methods to significantly enhance their efficacy and improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Davodabadi F, Sarhadi M, Arabpour J, Sargazi S, Rahdar A, Díez-Pascual AM. Breast cancer vaccines: New insights into immunomodulatory and nano-therapeutic approaches. J Control Release 2022; 349:844-875. [PMID: 35908621 DOI: 10.1016/j.jconrel.2022.07.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
Breast cancer (BC) is known to be a highly heterogeneous disease that is clinically subdivided into four primary molecular subtypes, each having distinct morphology and clinical implications. These subtypes are principally defined by hormone receptors and other proteins involved (or not involved) in BC development. BC therapeutic vaccines [including peptide-based vaccines, protein-based vaccines, nucleic acid-based vaccines (DNA/RNA vaccines), bacterial/viral-based vaccines, and different immune cell-based vaccines] have emerged as an appealing class of cancer immunotherapeutics when used alone or combined with other immunotherapies. Employing the immune system to eliminate BC cells is a novel therapeutic modality. The benefit of active immunotherapies is that they develop protection against neoplastic tissue and readjust the immune system to an anti-tumor monitoring state. Such immunovaccines have not yet shown effectiveness for BC treatment in clinical trials. In recent years, nanomedicines have opened new windows to increase the effectiveness of vaccinations to treat BC. In this context, some nanoplatforms have been designed to efficiently deliver molecular, cellular, or subcellular vaccines to BC cells, increasing the efficacy and persistence of anti-tumor immunity while minimizing undesirable side effects. Immunostimulatory nano-adjuvants, liposomal-based vaccines, polymeric vaccines, virus-like particles, lipid/calcium/phosphate nanoparticles, chitosan-derived nanostructures, porous silicon microparticles, and selenium nanoparticles are among the newly designed nanostructures that have been used to facilitate antigen internalization and presentation by antigen-presenting cells, increase antigen stability, enhance vaccine antigenicity and remedial effectivity, promote antigen escape from the endosome, improve cytotoxic T lymphocyte responses, and produce humoral immune responses in BC cells. Here, we summarized the existing subtypes of BC and shed light on immunomodulatory and nano-therapeutic strategies for BC vaccination. Finally, we reviewed ongoing clinical trials on BC vaccination and highlighted near-term opportunities for moving forward.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran.
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran.
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
7
|
Barati N, Motavallihaghi S, Nikfar B, Chaichian S, Momtazi-Borojeni AA. Potential therapeutic effects of Ivermectin in COVID-19. Exp Biol Med (Maywood) 2022; 247:1388-1396. [PMID: 35686662 PMCID: PMC9442455 DOI: 10.1177/15353702221099579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
COVID-19 is a critical pandemic that affected communities around the world, and there is currently no specific drug treatment for it. The virus enters the human cells via spikes and induces cytokine production and finally arrests the cell cycle. Ivermectin shows therapeutic potential for treating COVID-19 infection based on in vitro studies. Docking studies have shown a strong affinity between Ivermectin and some virulence factors of COVID-19. Notably, clinical evidence has demonstrated that Ivermectin with usual doses is effective by both the prophylactic and therapeutic approaches in all phases of the disease. Ivermectin inhibits both the adhesion and replication of the virus. Local therapy of the lung with Ivermectin or combination therapy may get better results and decrease the dose of the drug.
Collapse
Affiliation(s)
- Nastaran Barati
- Research Center For Molecular
Medicine, Hamadan University of Medical Sciences, Hamadan 9174223425,
Iran
- Medicinal Plants and Natural
Products Research Center, Hamadan University of Medical Sciences, Hamadan
9174223425, Iran
| | | | - Banafsheh Nikfar
- Pars Advanced and Minimally
Invasive Medical Manners Research Center, Pars Hospital, Iran University of
Medical Sciences, Tehran 1415944911, Iran
| | - Shahla Chaichian
- Pars Advanced and Minimally
Invasive Medical Manners Research Center, Pars Hospital, Iran University of
Medical Sciences, Tehran 1415944911, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical
Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences,
Mashhad 8167994434, Iran
| |
Collapse
|
8
|
Ahmad MZ, Alasiri AS, Alasmary MY, Abdullah MM, Ahmad J, Abdel Wahab BA, M Alqahtani SA, Pathak K, Mustafa G, Khan MA, Saikia R, Gogoi U. Emerging advances in nanomedicine for breast cancer immunotherapy: opportunities and challenges. Immunotherapy 2022; 14:957-983. [PMID: 35852105 DOI: 10.2217/imt-2021-0348] [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] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is one of the most common causes of cancer-related morbidity and mortality in women worldwide. Early diagnosis and an appropriate therapeutic approach for all cancers are climacterics for a favorable prognosis. Targeting the immune system in breast cancer is already a clinical reality with notable successes, specifically with checkpoint blockade antibodies and chimeric antigen receptor T-cell therapy. However, there have been inevitable setbacks in the clinical application of cancer immunotherapy, including inadequate immune responses due to insufficient delivery of immunostimulants to immune cells and uncontrolled immune system modulation. Rapid advancements and new evidence have suggested that nanomedicine-based immunotherapy may be a viable option for treating breast cancer.
Collapse
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Ali S Alasiri
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Mohammed Yahia Alasmary
- Medical Department, College of Medicine, Najran University, Najran, 11001, Kingdom of Saudi Arabia
| | - M M Abdullah
- Advanced Materials & Nano-Research Centre, Department of Physics, Faculty of Science & Arts, Najran University, Najran, 11001, Kingdom Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Basel A Abdel Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, 11001, Kingdom of Saudi Arabia
- Department of Pharmacology, College of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Saif Aboud M Alqahtani
- Internal Medicine Department, College of Medicine, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
| | - Kalyani Pathak
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Gulam Mustafa
- College of Pharmacy, Shaqra University, Ad-Dawadmi Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Ahmad Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Riya Saikia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Urvashee Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| |
Collapse
|
9
|
Potent anti-tumor immune response and tumor growth inhibition induced by HER2 subdomain fusion protein in a mouse tumor model. J Cancer Res Clin Oncol 2022; 149:2437-2450. [PMID: 35737089 DOI: 10.1007/s00432-022-04084-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/20/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE Several approaches have so far been employed to establish anti-tumor immunity by targeting HER2 protein. Active immunization with recombinant HER2 subdomains has previously been demonstrated to induce potent immune response and tumor growth inhibition. In the present study, we investigated the immunogenicity and tumor inhibitory effect of a fusion protein consisting of human HER2 extracellular subdomain (ECD-DI + II) together with T-helper cell epitopes of Tetanus toxin (p2 and p30). METHODS BALB/c mice were immunized with two recombinant proteins (DI + II and p2p30-DI + II) emulsified in 4 different adjuvants. Anti-DI + II antibody response, cytokine profile, frequency of splenic CD25+FOXP3+ regulatory T cells (Tregs) and CD8+CD107a+ cytotoxic T lymphocytes (CTLs) were assessed in the immunized mice. To assess the anti-tumor effect, the immunized mice were subcutaneously challenged with HER2-overexpressing tumor cells and the tumor growth was determined. RESULTS Both recombinant proteins were able to induce comparable levels of ECD-DI + II-specific antibodies. Immunization with p2p30-DI + II resulted in a significant increase in the level of Interferon-gamma (IFN-γ) secretion compared to DI + II protein and significantly higher frequency of CTLs and lower frequency of Tregs. The number of mice that remained tumor-free until day 120 was significantly higher in p2p30-DI + II vaccinated groups. CONCLUSIONS Our data suggest that the p2p30-DI + II fusion protein together with CpG adjuvant induces more potent anti-tumor immune responses in a mouse tumor model. Accordingly, this formulation might be considered as a potential immunotherapeutic approach in HER2+ cancers.
Collapse
|
10
|
Behravan N, Zahedipour F, Jaafari MR, Johnston TP, Sahebkar A. Lipid-based nanoparticulate delivery systems for HER2-positive breast cancer immunotherapy. Life Sci 2022; 291:120294. [PMID: 34998838 DOI: 10.1016/j.lfs.2021.120294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Lipid-based nanoparticulate delivery platforms such as liposomes help overcome cell and tissue barriers and allow prolonged therapeutic plasma drug concentrations, simultaneous targeting of tumor tissue, and increased bioavailability of numerous drugs used for treatment of cancer. The human epidermal growth factor receptor, HER2, is an important player in the pathogenesis of breast cancer and is considered a potential cancer biomarker for the design of immunotherapeutics. HER2-positive breast cancer is found in up to 30% of breast cancer patients. Currently, a variety of lipid nanoparticulate systems are being evaluated in preclinical settings and in clinical trials for targeting HER2-positive breast cancer. Advances in functionalized anti-HER2 lipid nanoparticulates have demonstrated promise and may lead to the development of new nano-immunotherapy protocols against HER2 positive breast cancer. Here we present a review of the most up-to-date literature, including our own research, on the use of lipid nanoparticulate carriers in immunotherapy of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Nima Behravan
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Barati N, Nikpoor AR, Mosaffa F, Razazan A, Badiee A, Motavallihaghi SM, Behravan J, Jaafari MR. AE36 HER2/neu-derived peptide linked to positively charged liposomes with CpG-ODN as an effective therapeutic and prophylactic vaccine for breast cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
12
|
Fobian SF, Cheng Z, ten Hagen TLM. Smart Lipid-Based Nanosystems for Therapeutic Immune Induction against Cancers: Perspectives and Outlooks. Pharmaceutics 2021; 14:26. [PMID: 35056922 PMCID: PMC8779430 DOI: 10.3390/pharmaceutics14010026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy, a promising and widely applied mode of oncotherapy, makes use of immune stimulants and modulators to overcome the immune dysregulation present in cancer, and leverage the host's immune capacity to eliminate tumors. Although some success has been seen in this field, toxicity and weak immune induction remain challenges. Liposomal nanosystems, previously used as targeting agents, are increasingly functioning as immunotherapeutic vehicles, with potential for delivery of contents, immune induction, and synergistic drug packaging. These systems are tailorable, multifunctional, and smart. Liposomes may deliver various immune reagents including cytokines, specific T-cell receptors, antibody fragments, and immune checkpoint inhibitors, and also present a promising platform upon which personalized medicine approaches can be built, especially with preclinical and clinical potentials of liposomes often being frustrated by inter- and intrapatient variation. In this review, we show the potential of liposomes in cancer immunotherapy, as well as the methods for synthesis and in vivo progression thereof. Both preclinical and clinical studies are included to comprehensively illuminate prospects and challenges for future research and application.
Collapse
Affiliation(s)
| | | | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology (LEO), Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (S.-F.F.); (Z.C.)
| |
Collapse
|