1
|
Farooq MA, Johnston APR, Trevaskis NL. Impact of nanoparticle properties on immune cell interactions in the lymph node. Acta Biomater 2024:S1742-7061(24)00758-X. [PMID: 39701340 DOI: 10.1016/j.actbio.2024.12.039] [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: 08/07/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
The lymphatic system plays an important role in health and many diseases, such as cancer, autoimmune, cardiovascular, metabolic, hepatic, viral, and other infectious diseases. The lymphatic system is, therefore, an important treatment target site for a range of diseases. Lymph nodes (LNs), rich in T cells, B cells, dendritic cells, and macrophages, are also primary sites of action for vaccines and immunotherapies. Promoting the delivery of therapeutics and vaccines to LNs can, therefore, enhance treatment efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. Several nanoparticle (NP) based delivery systems, such as polymeric NPs, lipid NPs, liposomes, micelles, and dendrimers, have been reported to enhance the delivery of therapeutics and/or vaccines to LNs. Specific uptake into the lymph following injection into tissues is highly dependent on particle properties, particularly particle size, as small molecules are more likely to be taken up by blood capillaries due to higher blood flow rates, whereas larger molecules and NPs can be specifically transported via the lymphatic vessels to LNs as the initial lymphatic capillaries are more permeable than blood capillaries. Once NPs enter LNs, particle properties also have an important influence on their disposition within the node and association with immune cells, which has significant implications for the design of vaccines and immunotherapies. This review article focuses on the impact of NP properties, such as size, surface charge and modification, and route of administration, on lymphatic uptake, retention, and interactions with immune cells in LNs. We suggest that optimizing all these factors can enhance the efficacy of vaccines or therapeutics with targets in the lymphatics and also be helpful for the rational design of vaccines. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an essential role in health and is an important treatment target site for a range of diseases. Promoting the delivery of immunotherapies and vaccines to immune cells in lymph nodes can enhance efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. One of the major approaches used to deliver therapeutics and vaccines to lymph nodes is via injection in nanoparticle delivery systems. This review aims to provide an overview of the impact of nanoparticle properties, such as size, surface charge, modification, and route of administration, on lymphatic uptake, lymph node retention, and interactions with immune cells in lymph nodes. This will inform the design of future improved nanoparticle systems for vaccines and immunotherapies.
Collapse
Affiliation(s)
- Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
2
|
Cheng S, Long X, Zhang Y, Lan X, Jiang D. Advancing Cancer Vaccines with Radionuclide Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2406950. [PMID: 39530610 DOI: 10.1002/smll.202406950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Cancer vaccines are emerged as a beacon of hope in the fight against cancer. However, the lack of effective methods to directly observe their in vivo behavior and monitor therapeutic responses hinders their translation into clinical settings. Radionuclide imaging allows for non-invasive and real-time visualization of vaccine biodistribution and immunological response, offering valuable insights into the effectiveness of cancer vaccines and aiding in patient stratification. In this review, the latest advances and potential applications of radionuclide imaging in cancer vaccines are discussed, with a specific focus on strategies for visualizing the spatiotemporal distribution of vaccines in vivo and monitoring treatment efficacy. The challenges and considerations for implementing these techniques in clinical practice are also highlighted, aiming to inform and guide future research in this field.
Collapse
Affiliation(s)
- Sixuan Cheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xingru Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yongxue Zhang
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaoli Lan
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Dawei Jiang
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| |
Collapse
|
3
|
Chen Y, Li Z, Wei Y, Guo X, Li M, Xia Y, Wu Y, Liao M, Wang S, Wang H, Zhou X, Lan F, Cheng L. Effects of a Novel Magnetic Nanomaterial on Oral Biofilms. Int Dent J 2024:S0020-6539(24)01412-6. [PMID: 39322517 DOI: 10.1016/j.identj.2024.07.1219] [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: 04/22/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 09/27/2024] Open
Abstract
Dental caries is one of the most common oral chronic infectious diseases, and novel antibacterial materials must be developed to control plaque and inhibit formation of dental caries. Combining magnetic nanomaterials with antibacterial agents to decrease the formation of bacterial biofilm has been a hot topic in the biomedical field. The present study developed a novel magnetic nanomaterial chemically combined with dimethylaminododecyl methacrylate (DMADDM) and initially investigated its inhibiting effects on biofilms by using traditional caries-related bacteria and saliva flora models. The novel magnetic nanomaterials successfully loaded DMADDM according to thermogravimetric analysis, Fourier transform infrared spectroscopy, x-ray diffraction, vibrating sample magnetometry, scanning electron microscopy, and transmission electron microscopy results. Further, the novel nanoparticle Fe3O4@SiO2@DMADDM with concentration of 8 mg/mL could effectively reduce Streptococcus mutans biofilm and decrease the production of lactic acid. The 16S rDNA sequencing revealed that Fe3O4@SiO2@DMADDM could depress the proportion of caries-related bacteria in saliva-derived biofilm, such as Streptococcus, Veillonella, and Neisseria. Therefore, Fe3O4@SiO2@DMADDM is a novel effective antibacterial magnetic nanomaterial and has clinical potential in plaque control and dental caries prevention.
Collapse
Affiliation(s)
- Yanru Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zhiyu Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Yu Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiao Guo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Mingyun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Min Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Suping Wang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haohao Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Fang Lan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Qiu J, Jiang Y, Ye N, Jin G, Shi H, Qian D. Leveraging the intratumoral microbiota to treat human cancer: are engineered exosomes an effective strategy? J Transl Med 2024; 22:728. [PMID: 39103887 DOI: 10.1186/s12967-024-05531-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Cancer remains a leading cause of global mortality. The tumor microbiota has increasingly been recognized as a key regulator of cancer onset and progression, in addition to shaping tumor responses to immunotherapy. Microbes, including viruses, bacteria, fungi, and other eukaryotic species can impact the internal homeostasis and health of humans. Research focused on the gut microflora and the intratumoral microbiome has revolutionized the current understanding of how tumors grow, progress, and resist therapeutic interventions. Even with this research, however, there remains relatively little that is known with respect to the abundance of microbes and their effects on tumors and the tumor microenvironment. Engineered exosomes are a class of artificial extracellular nanovesicles that can actively transport small molecule drugs and nucleic acids, which have the broad prospects of tumor cell therapy. The present review offers an overview of recent progress and challenges associated with the intratumoral microbiome and engineered exosomes in the context of cancer research. These discussions are used to inform the construction of a novel framework for engineered exosome-mediated targeted drug delivery, taking advantage of intratumoral microbiota diversity as a strategic asset and thereby providing new opportunities to more effectively treat and manage cancer in the clinic.
Collapse
Affiliation(s)
- Jie Qiu
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Yuancong Jiang
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Nanwei Ye
- Department of Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Gan Jin
- Department of Vascular Hernia Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Hao Shi
- Department of Radiotherapy, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Da Qian
- Department of Burn and Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, Jiangsu Province, 215500, China
- Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People ' s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China
| |
Collapse
|
5
|
Chattopadhyay S, Hazra R, Mallick A, Gayen S, Roy S. A review on comprehending immunotherapeutic approaches inducing ferroptosis: Managing tumour immunity. Immunology 2024; 172:547-565. [PMID: 38566448 DOI: 10.1111/imm.13789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
Ferroptosis, a necrotic, iron-dependent controlled cell death mechanism, is distinguished by the development of lipid peroxides to fatal proportions. Malignant tumours, influenced by iron to promote fast development, are vulnerable to ferroptosis. Based upon mounting evidence it has been observed that ferroptosis may be immunogenic and hence may complement immunotherapies. A new approach includes iron oxide-loaded nano-vaccines (IONVs), having supremacy for the traits of the tumour microenvironment (TME) to deliver specific antigens through improving the immunostimulatory capacity by molecular disintegration and reversible covalent bonds that target the tumour cells and induce ferroptosis. Apart from IONVs, another newer approach to induce ferroptosis in tumour cells is through oncolytic virus (OVs). One such oncolytic virus is the Newcastle Disease Virus (NDV), which can only multiply in cancer cells through the p53-SLC7A11-GPX4 pathway that leads to elevated levels of lipid peroxide and intracellular reactive oxygen species leading to the induction of ferroptosis that induce ferritinophagy.
Collapse
Affiliation(s)
- Soumyadeep Chattopadhyay
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal, India
| | - Rudradeep Hazra
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal, India
| | - Arijit Mallick
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal, India
| | - Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal, India
| |
Collapse
|
6
|
Ahmad I, Altameemi KKA, Hani MM, Ali AM, Shareef HK, Hassan ZF, Alubiady MHS, Al-Abdeen SHZ, Shakier HG, Redhee AH. Shifting cold to hot tumors by nanoparticle-loaded drugs and products. Clin Transl Oncol 2024:10.1007/s12094-024-03577-3. [PMID: 38922537 DOI: 10.1007/s12094-024-03577-3] [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: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Cold tumors lack antitumor immunity and are resistant to therapy, representing a major challenge in cancer medicine. Because of the immunosuppressive spirit of the tumor microenvironment (TME), this form of tumor has a low response to immunotherapy, radiotherapy, and also chemotherapy. Cold tumors have low infiltration of immune cells and a high expression of co-inhibitory molecules, such as immune checkpoints and immunosuppressive molecules. Therefore, targeting TME and remodeling immunity in cold tumors can improve the chance of tumor repression after therapy. However, tumor stroma prevents the infiltration of inflammatory cells and hinders the penetration of diverse molecules and drugs. Nanoparticles are an intriguing tool for the delivery of immune modulatory agents and shifting cold to hot tumors. In this review article, we discuss the mechanisms underlying the ability of nanoparticles loaded with different drugs and products to modulate TME and enhance immune cell infiltration. We also focus on newest progresses in the design and development of nanoparticle-based strategies for changing cold to hot tumors. These include the use of nanoparticles for targeted delivery of immunomodulatory agents, such as cytokines, small molecules, and checkpoint inhibitors, and for co-delivery of chemotherapy drugs and immunomodulatory agents. Furthermore, we discuss the potential of nanoparticles for enhancing the efficacy of cancer vaccines and cell therapy for overcoming resistance to treatment.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | | | - Mohaned Mohammed Hani
- Department of Medical Instrumentation Engineering Techniques, Imam Ja'afar Al-Sadiq University, Al Muthanna, Iraq
| | - Afaq Mahdi Ali
- Department of Pharmaceutics, Al-Turath University College, Baghdad, Iraq
| | - Hasanain Khaleel Shareef
- Department of Medical Biotechnology, College of Science, Al-Mustaqbal University, Hilla, Iraq
- Biology Department, College of Science for Women, University of Babylon, Hilla, Iraq
| | | | | | | | | | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
7
|
Park D, Lee SJ, Park JW. Aptamer-Based Smart Targeting and Spatial Trigger-Response Drug-Delivery Systems for Anticancer Therapy. Biomedicines 2024; 12:187. [PMID: 38255292 PMCID: PMC10813750 DOI: 10.3390/biomedicines12010187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
In recent years, the field of drug delivery has witnessed remarkable progress, driven by the quest for more effective and precise therapeutic interventions. Among the myriad strategies employed, the integration of aptamers as targeting moieties and stimuli-responsive systems has emerged as a promising avenue, particularly in the context of anticancer therapy. This review explores cutting-edge advancements in targeted drug-delivery systems, focusing on the integration of aptamers and stimuli-responsive platforms for enhanced spatial anticancer therapy. In the aptamer-based drug-delivery systems, we delve into the versatile applications of aptamers, examining their conjugation with gold, silica, and carbon materials. The synergistic interplay between aptamers and these materials is discussed, emphasizing their potential in achieving precise and targeted drug delivery. Additionally, we explore stimuli-responsive drug-delivery systems with an emphasis on spatial anticancer therapy. Tumor microenvironment-responsive nanoparticles are elucidated, and their capacity to exploit the dynamic conditions within cancerous tissues for controlled drug release is detailed. External stimuli-responsive strategies, including ultrasound-mediated, photo-responsive, and magnetic-guided drug-delivery systems, are examined for their role in achieving synergistic anticancer effects. This review integrates diverse approaches in the quest for precision medicine, showcasing the potential of aptamers and stimuli-responsive systems to revolutionize drug-delivery strategies for enhanced anticancer therapy.
Collapse
Affiliation(s)
- Dongsik Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Su Jin Lee
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Jee-Woong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| |
Collapse
|
8
|
Priyanka, Abusalah MAH, Chopra H, Sharma A, Mustafa SA, Choudhary OP, Sharma M, Dhawan M, Khosla R, Loshali A, Sundriyal A, Saini J. Nanovaccines: A game changing approach in the fight against infectious diseases. Biomed Pharmacother 2023; 167:115597. [PMID: 37783148 DOI: 10.1016/j.biopha.2023.115597] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
The field of nanotechnology has revolutionised global attempts to prevent, treat, and eradicate infectious diseases in the foreseen future. Nanovaccines have proven to be a valuable pawn in this novel technology. Nanovaccines are made up of nanoparticles that are associated with or prepared with components that can stimulate the host's immune system. In addition to their delivery capabilities, the nanocarriers have been demonstrated to possess intrinsic adjuvant properties, working as immune cell stimulators. Thus, nanovaccines have the potential to promote rapid as well as long-lasting humoral and cellular immunity. The nanovaccines have several possible benefits, including site-specific antigen delivery, increased antigen bioavailability, and a diminished adverse effect profile. To avail these benefits, several nanoparticle-based vaccines are being developed, including virus-like particles, liposomes, polymeric nanoparticles, nanogels, lipid nanoparticles, emulsion vaccines, exomes, and inorganic nanoparticles. Inspired by their distinctive properties, researchers are working on the development of nanovaccines for a variety of applications, such as cancer immunotherapy and infectious diseases. Although a few challenges still need to be overcome, such as modulation of the nanoparticle pharmacokinetics to avoid rapid elimination from the bloodstream by the reticuloendothelial system, The future prospects of this technology are also assuring, with multiple options such as personalised vaccines, needle-free formulations, and combination nanovaccines with several promising candidates.
Collapse
Affiliation(s)
- Priyanka
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India
| | - Mai Abdel Haleem Abusalah
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Abhilasha Sharma
- Department of Life Science, Gujarat University, University School of Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Suhad Asad Mustafa
- Scientific Research Center/ Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India.
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, Punjab, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, Punjab, India; Trafford College, Altrincham, Manchester WA14 5PQ, UK.
| | - Rajiv Khosla
- Department of Biotechnology, Doaba College, Jalandhar 144004, Punjab, India
| | - Aanchal Loshali
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ankush Sundriyal
- School of Pharmaceutical Sciences and Research, Sardar Bhagwan Singh University, Balawala, Dehradun 248001, India
| | - Jyoti Saini
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India
| |
Collapse
|
9
|
Hasan N, Nadaf A, Imran M, Jiba U, Sheikh A, Almalki WH, Almujri SS, Mohammed YH, Kesharwani P, Ahmad FJ. Skin cancer: understanding the journey of transformation from conventional to advanced treatment approaches. Mol Cancer 2023; 22:168. [PMID: 37803407 PMCID: PMC10559482 DOI: 10.1186/s12943-023-01854-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/30/2023] [Indexed: 10/08/2023] Open
Abstract
Skin cancer is a global threat to the healthcare system and is estimated to incline tremendously in the next 20 years, if not diagnosed at an early stage. Even though it is curable at an early stage, novel drug identification, clinical success, and drug resistance is another major challenge. To bridge the gap and bring effective treatment, it is important to understand the etiology of skin carcinoma, the mechanism of cell proliferation, factors affecting cell growth, and the mechanism of drug resistance. The current article focusses on understanding the structural diversity of skin cancers, treatments available till date including phytocompounds, chemotherapy, radiotherapy, photothermal therapy, surgery, combination therapy, molecular targets associated with cancer growth and metastasis, and special emphasis on nanotechnology-based approaches for downregulating the deleterious disease. A detailed analysis with respect to types of nanoparticles and their scope in overcoming multidrug resistance as well as associated clinical trials has been discussed.
Collapse
Affiliation(s)
- Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Arif Nadaf
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Imran
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, 4102, Australia
| | - Umme Jiba
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, 24381, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, 61421, Asir-Abha, Saudi Arabia
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Kuthambakkam, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
10
|
Liu M, Zhao Y, Shi Z, Zink JI, Yu Q. Virus-like Magnetic Mesoporous Silica Particles as a Universal Vaccination Platform against Pathogenic Infections. ACS NANO 2023; 17:6899-6911. [PMID: 36961475 DOI: 10.1021/acsnano.3c00644] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Vaccination is the most important way of population protection from life-threatening pathogenic infections. However, its efficiency is frequently compromised by a failure of strong antigen presentation and immune activation. Herein, we developed virus-like magnetic mesoporous silica nanoparticles as a universal vaccination platform (termed MagParV) for preventing pathogenic infections. This platform was constructed by integrating synthetic biology-based endoplasmic reticulum-targeting vesicles with magnetic mesoporous silica particles. This platform exhibited high antigen-loading capacity, strongly targeting the endoplasmic reticulum and promoting antigen presentation in dendritic cells. After prime-boost vaccination, the antigen-loading MagParV with AMF drastically elicited specific antibody production against corresponding antigens of fungal, bacterial, and viral pathogens. A systemic infection model further revealed that the platform effectively protected the mice from severe fungal systemic infections. This study realized synthetic biology-facilitated green manufacturing of vaccines, which is promising for magnetism-activated vaccination against different kinds of pathogenic infections.
Collapse
Affiliation(s)
- Mingyang Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, People's Republic of China
| | - Yan Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Zhishang Shi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- California Nano Systems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
11
|
Yan H, Lin G, Liu Z, Gu F, Zhang Y. Nano-adjuvants and immune agonists promote antitumor immunity of peptide amphiphiles. Acta Biomater 2023; 161:213-225. [PMID: 36858163 DOI: 10.1016/j.actbio.2023.02.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/04/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Immunostimulatory cues play an important role in priming antitumor immunity and promoting the efficacy of subunit cancer vaccines. However, the clinical use of many immunostimulatory agents is often hampered by their inefficient in vivo delivery which may decrease immune response to the vaccination. To promote vaccine efficacy, we develop vaccine formulations which integrate three key elements: (1) a nano-adjuvant formulated by conjugating an agonistic anti-CD40 monoclonal antibody (αCD40) to the surface of a polyIC-loaded lipid nanoparticle, (2) a peptide amphiphile containing an optimized CD8+ T-cell epitope that derived from a melanoma antigen gp100, (3) an agonistic anti-4-1BB monoclonal antibody (α4-1BB) that boosts the efficacy of vaccinations. In a syngeneic mouse model of melanoma, the vaccine formulations enhanced innate immunity and activated multiple innate immune signaling pathways within draining lymph nodes, as well as promoted antigen-specific immune responses and reduced immunosuppression in the tumor microenvironment, leading to profound tumor growth inhibition and prolonged survival. Thus, our vaccine formulations represent an attractive strategy to stimulate antitumor immunity and control tumor progression. STATEMENT OF SIGNIFICANCE: The clinical use of many immunostimulatory agents is often hampered by their inefficient in vivo delivery which may decrease immune response to the vaccination. To promote the antitumor immunity of subunit vaccines, we develop novel vaccine formulations that integrate multifunctional modalities including (1) a nano-adjuvant containing anti-CD40 monoclonal antibody (αCD40) and TLR3 agonist which activate innate immunity through diverse signaling pathways, (2) a peptide amphiphile containing an optimized CD8+ T-cell epitope from tumor antigen, (3) an anti-4-1BB monoclonal antibody (α4-1BB) that boosts the efficacy of vaccinations. In this study, our vaccine formulations stimulate superior antitumor immunity and control tumor progression. The above nano-engineered platform and immunogenic biomacromolecules can be further applied to other T-cell-inducing vaccines.
Collapse
Affiliation(s)
- Huan Yan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Guibin Lin
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhanyan Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Fei Gu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
12
|
Khizar S, Elkalla E, Zine N, Jaffrezic-Renault N, Errachid A, Elaissari A. Magnetic nanoparticles: multifunctional tool for cancer therapy. Expert Opin Drug Deliv 2023; 20:189-204. [PMID: 36608938 DOI: 10.1080/17425247.2023.2166484] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Cancer has one of the highest mortality rates globally. The traditional therapies used to treat cancer have harmful adverse effects. Considering these facts, researchers have explored new therapeutic possibilities with enhanced benefits. Nanoparticle development for cancer detection, in addition to therapy, has shown substantial progress over the past few years. AREA COVERED Herein, the latest research regarding cancer treatment employing magnetic nanoparticles (MNPs) in chemo-, immuno-, gene-, and radiotherapy along with hyperthermia is summarized, in addition to their physio-chemical features, advantages, and limitations for clinical translation have also been discussed. EXPERT OPINION MNPs are being extensively investigated and developed into effective modules for cancer therapy. They are highly functional tools aimed at cancer therapy owing to their excellent superparamagnetic, chemical, biocompatible, physical, and biodegradable properties.
Collapse
Affiliation(s)
- Sumera Khizar
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | - Eslam Elkalla
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | - Nadia Zine
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | | | - Abdelhamid Errachid
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | | |
Collapse
|
13
|
Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm (Beijing) 2023; 4:e187. [PMID: 36654533 PMCID: PMC9834710 DOI: 10.1002/mco2.187] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2023] Open
Abstract
Cancer is a complex disease associated with a combination of abnormal physiological process and exhibiting dysfunctions in multiple systems. To provide effective treatment and diagnosis for cancer, current treatment strategies simultaneously focus on various tumor targets. Based on the rapid development of nanotechnology, nanocarriers have been shown to exhibit excellent potential for cancer therapy. Compared with nanoparticles with single functions, multifunctional nanoparticles are believed to be more aggressive and potent in the context of tumor targeting. However, the development of multifunctional nanoparticles is not simply an upgraded version of the original function, but involves a sophisticated system with a proper backbone, optimized modification sites, simple preparation method, and efficient function integration. Despite this, many well-designed multifunctional nanoparticles with promising therapeutic potential have emerged recently. Here, to give a detailed understanding and analyzation of the currently developed multifunctional nanoparticles, their platform structures with organic or inorganic backbones were systemically generalized. We emphasized on the functionalization and modification strategies, which provide additional functions to the nanoparticle. We also discussed the application combination strategies that were involved in the development of nanoformulations with functional crosstalk. This review thus provides an overview of the construction strategies and application advances of multifunctional nanoparticles.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Xingmei Duan
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuan ProvinceChina
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
14
|
Wu Y, Zhang Z, Wei Y, Qian Z, Wei X. Nanovaccines for cancer immunotherapy: Current knowledge and future perspectives. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.108098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
15
|
Wang N, Zuo Y, Wu S, Huang C, Zhang L, Zhu D. Spatio-temporal delivery of both intra- and extracellular toll-like receptor agonists for enhancing antigen-specific immune responses. Acta Pharm Sin B 2022; 12:4486-4500. [PMID: 36561992 PMCID: PMC9764069 DOI: 10.1016/j.apsb.2022.05.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 12/25/2022] Open
Abstract
For cancer immunotherapy, triggering toll-like receptors (TLRs) in dendritic cells (DCs) can potentiate antigen-based immune responses. Nevertheless, to generate robust and long-lived immune responses, a well-designed nanovaccine should consider different locations of TLRs on DCs and co-deliver both antigens and TLR agonist combinations to synergistically induce optimal antitumor immunity. Herein, we fabricated lipid-polymer hybrid nanoparticles (LPNPs) to spatio-temporally deliver model antigen ovalbumin (OVA) on the surface of the lipid layer, TLR4 agonist monophosphoryl lipid A (MPLA) within the lipid layer, and TLR7 agonist imiquimod (IMQ) in the polymer core to synergistically activate DCs by both extra- and intra-cellular TLRs for enhancing adaptive immune responses. LPNPs-based nanovaccines exhibited a narrow size distribution at the mean diameter of 133.23 nm and zeta potential of -2.36 mV, showed a high OVA loading (around 70.83 μg/mg) and IMQ encapsulation efficiency (88.04%). Our data revealed that LPNPs-based nanovaccines showed great biocompatibility to immune cells and an excellent ability to enhance antigen internalization, thereby promoting DCs maturation and cytokines production. Compared to Free OVA, OVA-LPNPs promoted antigen uptake, lysosome escape, depot effect and migration to secondary lymphatic organs. In vivo immunization showed that IMQ-MPLA-OVA-LPNPs with dual agonists induced more powerful cellular and humoral immune responses. Moreover, prophylactic vaccination by IMQ-MPLA-OVA-LPNPs effectively suppressed tumor growth and increased survival efficacy. Hence, the nanovaccines we fabricated can effectively co-deliver antigens and different TLR agonists and realize coordinated stimulation of DCs in a spatio-temporal manner for enhanced immune responses, which provides a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Nannan Wang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yueyue Zuo
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Shengjie Wu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Chenlu Huang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Linhua Zhang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
16
|
Yang Y, Li H, Fotopoulou C, Cunnea P, Zhao X. Toll-like receptor-targeted anti-tumor therapies: Advances and challenges. Front Immunol 2022; 13:1049340. [PMID: 36479129 PMCID: PMC9721395 DOI: 10.3389/fimmu.2022.1049340] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors, originally discovered to stimulate innate immune reactions against microbial infection. TLRs also play essential roles in bridging the innate and adaptive immune system, playing multiple roles in inflammation, autoimmune diseases, and cancer. Thanks to the immune stimulatory potential of TLRs, TLR-targeted strategies in cancer treatment have proved to be able to regulate the tumor microenvironment towards tumoricidal phenotypes. Quantities of pre-clinical studies and clinical trials using TLR-targeted strategies in treating cancer have been initiated, with some drugs already becoming part of standard care. Here we review the structure, ligand, signaling pathways, and expression of TLRs; we then provide an overview of the pre-clinical studies and an updated clinical trial watch targeting each TLR in cancer treatment; and finally, we discuss the challenges and prospects of TLR-targeted therapy.
Collapse
Affiliation(s)
- Yang Yang
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Hongyi Li
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Christina Fotopoulou
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Paula Cunnea
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xia Zhao
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Bilbao‐Asensio M, Ruiz‐de‐Angulo A, Arguinzoniz AG, Cronin J, Llop J, Zabaleta A, Michue‐Seijas S, Sosnowska D, Arnold JN, Mareque‐Rivas JC. Redox‐Triggered Nanomedicine via Lymphatic Delivery: Inhibition of Melanoma Growth by Ferroptosis Enhancement and a Pt(IV)‐Prodrug Chemoimmunotherapy Approach. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Marc Bilbao‐Asensio
- Department of Chemistry Swansea University Singleton Park Swansea SA2 8PP UK
| | | | | | - James Cronin
- Swansea University Medical School Singleton Park Swansea SA2 8PP UK
| | - Jordi Llop
- Radiochemistry and Nuclear Imaging Laboratory CIC biomaGUNE Paseo Miramón 182 San Sebastián 20014 Spain
| | - Aintzane Zabaleta
- Clinica Universidad de Navarra Centro de Investigación Médica Aplicada (CIMA) IdiSNA Instituto de Investigación Sanitaria de Navarra Pamplona 31009 Spain
| | - Saul Michue‐Seijas
- Department of Chemistry Swansea University Singleton Park Swansea SA2 8PP UK
| | - Dominika Sosnowska
- School of Cancer and Pharmaceutical Sciences King's College London London SE1 1UL UK
| | - James N. Arnold
- School of Cancer and Pharmaceutical Sciences King's College London London SE1 1UL UK
| | | |
Collapse
|
18
|
Semple SL, Alkie TN, Jenik K, Warner BM, Tailor N, Kobasa D, DeWitte-Orr SJ. More tools for our toolkit: The application of HEL-299 cells and dsRNA-nanoparticles to study human coronaviruses in vitro. Virus Res 2022; 321:198925. [PMID: 36115551 PMCID: PMC9474404 DOI: 10.1016/j.virusres.2022.198925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 12/24/2022]
Abstract
Human coronaviruses (HCoVs) are important human pathogens, as exemplified by the current SARS-CoV-2 pandemic. While the ability of type I interferons (IFNs) to limit coronavirus replication has been established, the ability of double-stranded (ds)RNA, a potent IFN inducer, to inhibit coronavirus replication when conjugated to a nanoparticle is largely unexplored. Additionally, the number of IFN competent cell lines that can be used to study coronaviruses in vitro are limited. In the present study, we show that poly inosinic: poly cytidylic acid (pIC), when conjugated to a phytoglycogen nanoparticle (pIC+NDX) is able to protect IFN-competent human lung fibroblasts (HEL-299 cells) from infection with different HCoV species. HEL-299 was found to be permissive to HCoV-229E, -OC43 and MERS-CoV-GFP but not to HCoV-NL63 or SARS-CoV-2. Further investigation revealed that HEL-299 does not contain the required ACE2 receptor to enable propagation of both HCoV-NL63 and SARS-CoV-2. Following 24h exposure, pIC+NDX was observed to stimulate a significant, prolonged increase in antiviral gene expression (IFNβ, CXCL10 and ISG15) when compared to both NDX alone and pIC alone. This antiviral response translated into complete protection against virus production, for 4 days or 7 days post treatment with HCoV-229E or -OC43 when either pre-treated for 6h or 24h respectively. Moreover, the pIC+NDX combination also provided complete protection for 2d post infection when HEL-299 cells were infected with MERS-CoV-GFP following a 24h pretreatment with pIC+NDX. The significance of this study is two-fold. Firstly, it was revealed that HEL-299 cells can effectively be used as an IFN-competent model system for in vitro analysis of MERS-CoV. Secondly, pIC+NDX acts as a powerful inducer of type I IFNs in HEL-299, to levels that provide complete protection against coronavirus replication. This suggests an exciting and novel area of investigation for antiviral therapies that utilize innate immune stimulants. The results of this study will help to expand the range of available tools scientists have to investigate, and thus further understand, human coronaviruses.
Collapse
Affiliation(s)
- Shawna L Semple
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Tamiru N Alkie
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Kristof Jenik
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Bryce M Warner
- Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Nikesh Tailor
- Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
19
|
Systemic CD4 Immunity and PD-L1/PD-1 Blockade Immunotherapy. Int J Mol Sci 2022; 23:ijms232113241. [PMID: 36362027 PMCID: PMC9655397 DOI: 10.3390/ijms232113241] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
PD-L1/PD-1 blockade immunotherapy has changed the therapeutic approaches for the treatment of many cancers. Nevertheless, the mechanisms underlying its efficacy or treatment failure are still unclear. Proficient systemic immunity seems to be a prerequisite for efficacy, as recently shown in patients and in mouse models. It is widely accepted that expansion of anti-tumor CD8 T cell populations is principally responsible for anti-tumor responses. In contrast, the role of CD4 T cells has been less studied. Here we review and discuss the evidence supporting the contribution of CD4 T cells to anti-tumor immunity, especially recent advances linking CD4 T cell subsets to efficacious PD-L1/PD-1 blockade immunotherapy. We also discuss the role of CD4 T cell memory subsets present in peripheral blood before the start of immunotherapies, and their utility as predictors of response.
Collapse
|
20
|
Slezak AJ, Mansurov A, Raczy MM, Chang K, Alpar AT, Lauterbach AL, Wallace RP, Weathered RK, Medellin JE, Battistella C, Gray LT, Marchell TM, Gomes S, Swartz MA, Hubbell JA. Tumor Cell-Surface Binding of Immune Stimulating Polymeric Glyco-Adjuvant via Cysteine-Reactive Pyridyl Disulfide Promotes Antitumor Immunity. ACS CENTRAL SCIENCE 2022; 8:1435-1446. [PMID: 36313164 PMCID: PMC9615125 DOI: 10.1021/acscentsci.2c00704] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Indexed: 06/10/2023]
Abstract
Immune stimulating agents like Toll-like receptor 7 (TLR7) agonists induce potent antitumor immunity but are limited in their therapeutic window due to off-target immune activation. Here, we developed a polymeric delivery platform that binds excess unpaired cysteines on tumor cell surfaces and debris to adjuvant tumor neoantigens as an in situ vaccine. The metabolic and enzymatic dysregulation in the tumor microenvironment produces these exofacial free thiols, which can undergo efficient disulfide exchange with thiol-reactive pyridyl disulfide moieties upon intratumoral injection. These functional monomers are incorporated into a copolymer with pendant mannose groups and TLR7 agonists to target both antigen and adjuvant to antigen presenting cells. When tethered in the tumor, the polymeric glyco-adjuvant induces a robust antitumor response and prolongs survival of tumor-bearing mice, including in checkpoint-resistant B16F10 melanoma. The construct additionally reduces systemic toxicity associated with clinically relevant small molecule TLR7 agonists.
Collapse
Affiliation(s)
- Anna J. Slezak
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Aslan Mansurov
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Michal M. Raczy
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Kevin Chang
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Aaron T. Alpar
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Abigail L. Lauterbach
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Rachel P. Wallace
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Rachel K. Weathered
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Jorge E.G. Medellin
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Claudia Battistella
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Laura T. Gray
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Tiffany M. Marchell
- Committee
on Immunology, University of Chicago, Chicago, Illinois 60637, United States
| | - Suzana Gomes
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Melody A. Swartz
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Committee
on Immunology, University of Chicago, Chicago, Illinois 60637, United States
- Ben
May Department for Cancer Research, University
of Chicago, Chicago, Illinois 60637, United
States
- Committee
on Cancer Biology, University of Chicago, Chicago, Illinois 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Committee
on Immunology, University of Chicago, Chicago, Illinois 60637, United States
- Committee
on Cancer Biology, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
21
|
Andón FT, Leon S, Ummarino A, Redin E, Allavena P, Serrano D, Anfray C, Calvo A. Innate and Adaptive Responses of Intratumoral Immunotherapy with Endosomal Toll-Like Receptor Agonists. Biomedicines 2022; 10:1590. [PMID: 35884895 PMCID: PMC9313389 DOI: 10.3390/biomedicines10071590] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
Toll-like receptors (TLRs) are natural initial triggers of innate and adaptive immune responses. With the advent of cancer immunotherapy, nucleic acids engineered as ligands of endosomal TLRs have been investigated for the treatment of solid tumors. Despite promising results, their systemic administration, similarly to other immunotherapies, raises safety issues. To overcome these problems, recent studies have applied the direct injection of endosomal TLR agonists in the tumor and/or draining lymph nodes, achieving high local drug exposure and strong antitumor response. Importantly, intratumoral delivery of TLR agonists showed powerful effects not only against the injected tumors but also often against uninjected lesions (abscopal effects), resulting in some cases in cure and antitumoral immunological memory. Herein, we describe the structure and function of TLRs and their role in the tumor microenvironment. Then, we provide our vision on the potential of intratumor versus systemic delivery or vaccination approaches using TLR agonists, also considering the use of nanoparticles to improve their targeting properties. Finally, we collect the preclinical and clinical studies applying intratumoral injection of TLR agonists as monotherapies or in combination with: (a) other TLR or STING agonists; (b) other immunotherapies; (c) radiotherapy or chemotherapy; (d) targeted therapies.
Collapse
Affiliation(s)
- Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Sergio Leon
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
| | - Aldo Ummarino
- Laboratory of Cellular Immunology, Humanitas University, 20089 Pieve Emanuele, Italy; (A.U.); (C.A.)
| | - Esther Redin
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Paola Allavena
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
- Laboratory of Cellular Immunology, Humanitas University, 20089 Pieve Emanuele, Italy; (A.U.); (C.A.)
| | - Diego Serrano
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Clément Anfray
- Laboratory of Cellular Immunology, Humanitas University, 20089 Pieve Emanuele, Italy; (A.U.); (C.A.)
| | - Alfonso Calvo
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| |
Collapse
|
22
|
Zhu L, Mao H, Yang L. Advanced iron oxide nanotheranostics for multimodal and precision treatment of pancreatic ductal adenocarcinoma. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1793. [PMID: 35396932 PMCID: PMC9373845 DOI: 10.1002/wnan.1793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/22/2022] [Accepted: 03/10/2022] [Indexed: 06/14/2023]
Abstract
Despite current advances in new approaches for cancer detection and treatment, pancreatic cancer remains one of the most lethal cancer types. Difficult to detect early, aggressive tumor biology, and resistance to chemotherapy, radiotherapy, and immunotherapy result in a poor prognosis of pancreatic cancer patients with a 5-year survival of 10%. With advances in cancer nanotechnology, new imaging and drug delivery approaches that allow the development of multifunctional nanotheranostic agents offer opportunities for improving pancreatic cancer treatment using precision oncology. In this review, we will introduce potential applications of innovative theranostic strategies to address major challenges in the treatment of pancreatic cancer at different disease stages. Several important issues concerning targeted delivery of theranostic nanoparticles and tumor stromal barriers are discussed. We then focus on the development of a magnetic iron oxide nanoparticle platform for multimodal therapy of pancreatic cancer, including MRI monitoring targeted nanoparticle/drug delivery, therapeutic response, and tumor re-staging, activation of tumor immune response by immunoactivating nanoparticle and magnetic hyperthermia therapy, and intraoperative interventions for improving the outcome of targeted therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Atlanta, Georgia, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Atlanta, Georgia, USA
| |
Collapse
|
23
|
Nanomaterial-Based Drug Delivery System Targeting Lymph Nodes. Pharmaceutics 2022; 14:pharmaceutics14071372. [PMID: 35890268 PMCID: PMC9325242 DOI: 10.3390/pharmaceutics14071372] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/28/2022] [Accepted: 06/22/2022] [Indexed: 02/06/2023] Open
Abstract
The lymphatic system plays an indispensable role in humoral balance, lipid metabolism, and immune regulation. The lymph nodes (LNs) are known as the primary sites of tumor metastasis and the metastatic LNs largely affected the prognosis of the patiens. A well-designed lymphatic-targeted system favors disease treatment as well as vaccination efficacy. In recent years, development of nanotechnologies and emerging biomaterials have gained increasing attention in developing lymph-node-targeted drug-delivery systems. By mimicking the endogenous macromolecules or lipid conjugates, lymph-node-targeted nanocarries hold potential for disease diagnosis and tumor therapy. This review gives an introduction to the physiological functions of LNs and the roles of LNs in diseases, followed by a review of typical lymph-node-targeted nanomaterial-based drug-delivery systems (e.g., liposomes, micelles, inorganic nanomaterials, hydrogel, and nanocapsules). Future perspectives and conclusions concerned with lymph-node-targeted drug-delivery systems are also provided.
Collapse
|
24
|
Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression. Cancers (Basel) 2022; 14:cancers14122923. [PMID: 35740589 PMCID: PMC9221178 DOI: 10.3390/cancers14122923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Toll like receptors (TLRs) are a group of transmembrane receptors belonging to the class of pattern recognition receptors (PRR), which are involved in recognition of pathogen associated molecular patterns (PAMPs), inducing immune response. During the past decade, a number of preclinical and clinical breakthroughs in the field of TLR agonists has immerged in cancer research and some of these agents have performed exceptionally well in clinical trials. Based on evidence from scientific studies, we draw attention to several microbial based TLR agonists and discuss their relevance in various cancer and explore various microbial based TLR agonists for developing effective immunotherapeutic strategies against cancer. Abstract Toll-like receptors (TLRs) are typical transmembrane proteins, which are essential pattern recognition receptors in mediating the effects of innate immunity. TLRs recognize structurally conserved molecules derived from microbes and damage-associated molecular pattern molecules that play an important role in inflammation. Since the first discovery of the Toll receptor by the team of J. Hoffmann in 1996, in Drosophila melanogaster, numerous TLRs have been identified across a wide range of invertebrate and vertebrate species. TLR stimulation leads to NF-κB activation and the subsequent production of pro-inflammatory cytokines and chemokines, growth factors and anti-apoptotic proteins. The expression of TLRs has also been observed in many tumors, and their stimulation results in tumor progression or regression, depending on the TLR and tumor type. The anti-tumoral effects can result from the activation of anti-tumoral immune responses and/or the direct induction of tumor cell death. The pro-tumoral effects may be due to inducing tumor cell survival and proliferation or by acting on suppressive or inflammatory immune cells in the tumor microenvironment. The aim of this review is to draw attention to the effects of TLR stimulation in cancer, the activation of various TLRs by microbes in different types of tumors, and, finally, the role of TLRs in anti-cancer immunity and tumor rejection.
Collapse
|
25
|
Huang L, Ge X, Liu Y, Li H, Zhang Z. The Role of Toll-like Receptor Agonists and Their Nanomedicines for Tumor Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14061228. [PMID: 35745800 PMCID: PMC9230510 DOI: 10.3390/pharmaceutics14061228] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/20/2022] [Accepted: 06/07/2022] [Indexed: 01/11/2023] Open
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that play a critical role in innate and adaptive immunity. Toll-like receptor agonists (TLRa) as vaccine adjuvant candidates have become one of the recent research hotspots in the cancer immunomodulatory field. Nevertheless, numerous current systemic deliveries of TLRa are inappropriate for clinical adoption due to their low efficiency and systemic adverse reactions. TLRa-loaded nanoparticles are capable of ameliorating the risk of immune-related toxicity and of strengthening tumor suppression and eradication. Herein, we first briefly depict the patterns of TLRa, followed by the mechanism of agonists at those targets. Second, we summarize the emerging applications of TLRa-loaded nanomedicines as state-of-the-art strategies to advance cancer immunotherapy. Additionally, we outline perspectives related to the development of nanomedicine-based TLRa combined with other therapeutic modalities for malignancies immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Hui Li
- Correspondence: (H.L.); (Z.Z.)
| | | |
Collapse
|
26
|
Ren H, Li J, Liu G, Sun Y, Yang X, Jiang Z, Zhang J, Lovell JF, Zhang Y. Anticancer Vaccination with Immunogenic Micelles That Capture and Release Pristine CD8 + T-Cell Epitopes and Adjuvants. ACS APPLIED MATERIALS & INTERFACES 2022; 14:2510-2521. [PMID: 34986639 DOI: 10.1021/acsami.1c18117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The development of nanocarriers capable of codelivering antigens and immune-activating adjuvants is an emerging area of research and is relevant for cancer vaccines that target induction of antigen-specific CD8+ T-cell responses. Here, we report a system for delivery of short peptide antigens to dendritic cells for strong cellular immune responses, based on block copolymers chemically modified with a hydrophobic and self-immolative linker. After modification, micelles effectively and reversibly capture antigens and adjuvants via a covalent bond within several minutes in an aqueous solution. After uptake in antigen presenting cells, the polymer disulfide bond is cleaved by intracellular glutathione, leading to release of pristine antigens, along with the upregulated expression of costimulatory molecules. The induced antigen-specific CD8+ T cells have strong tumor cell killing efficacy in the murine B16OVA and human papilloma virus-E6/E7 subcutaneous and lung metastasis tumor models. In addition, delivery to lymph nodes can be imaged to visualize vaccine trafficking. Taken together, multifunctional self-immolative micelles represent a versatile class of a vaccine delivery system for the generation of a cellular immune response that warrants further exploration as a component of cancer immunotherapy.
Collapse
Affiliation(s)
- He Ren
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P. R. China 300350
| | - Jiexin Li
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P. R. China 300350
| | - Gengqi Liu
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P. R. China 300350
| | - Yaping Sun
- Imaging Center, Hospital of Traditional Chinese Medicine, Beichen District, Tianjin, P. R. China 300400
| | - Xingyue Yang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P. R. China 300350
| | - Zhen Jiang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P. R. China 300350
| | - Jingyu Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P. R. China 300350
| | - Jonathan F Lovell
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P. R. China 300350
| |
Collapse
|
27
|
Zhang BD, Wu JJ, Li WH, Hu HG, Zhao L, He PY, Zhao YF, Li YM. STING and TLR7/8 agonists-based nanovaccines for synergistic antitumor immune activation. NANO RESEARCH 2022; 15:6328-6339. [PMID: 35464625 PMCID: PMC9014842 DOI: 10.1007/s12274-022-4282-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 05/07/2023]
Abstract
UNLABELLED Immunostimulatory therapies based on pattern recognition receptors (PRRs) have emerged as an effective approach in the fight against cancer, with the ability to recruit tumor-specific lymphocytes in a low-immunogenicity tumor environment. The agonist cyclic dinucleotides (CDNs) of the stimulator of interferon gene (STING) are a group of very promising anticancer molecules that increase tumor immunogenicity by activating innate immunity. However, the tumor immune efficacy of CDNs is limited by several factors, including relatively narrow cytokine production, inefficient delivery to STING, and rapid clearance. In addition, a single adjuvant molecule is unable to elicit a broad cytokine response and thus cannot further amplify the anticancer effect. To address this problem, two or more agonist molecules are often used together to synergistically enhance immune efficacy. In this work, we found that a combination of the STING agonist CDGSF and the Toll-like receptor 7/8 (TLR7/8) agonist 522 produced a broader cytokine response. Subsequently, we developed multicomponent nanovaccines (MCNVs) consisting of a PC7A polymer as a nanocarrier encapsulating the antigen OVA and adjuvant molecules. These MCNVs activate bone marrow-derived dendritic cells (BMDCs) to produce multiple proinflammatory factors that promote antigen cross-presentation to stimulate specific antitumor T-cell responses. In in vivo experiments, we observed that MCNVs triggered a strong T-cell response in tumor-infiltrating lymphocytes, resulting in significant tumor regression and, notably, a 100% survival rate in mice through 25 days without other partnering therapies. These data suggest that our nanovaccines have great potential to advance cancer immunotherapy with increased durability and potency. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (synthesis of CDGSF, 522, PC7A and OVA; preparation of MCNVs; representative gating strategies for flow cytometry) is available in the online version of this article at 10.1007/s12274-022-4282-x.
Collapse
Affiliation(s)
- Bo-Dou Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Jun-Jun Wu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Hong-Guo Hu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Lang Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Pei-Yang He
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Yu-Fen Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315201 China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
- Beijing Institute for Brain Disorders, Beijing, 100069 China
| |
Collapse
|
28
|
Szulc-Kielbik I, Kielbik M. Tumor-Associated Macrophages: Reasons to Be Cheerful, Reasons to Be Fearful. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:107-140. [PMID: 35165862 DOI: 10.1007/978-3-030-91311-3_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor microenvironment (TME) is a complex and constantly evolving entity that consists not only of cancer cells, but also of resident host cells and immune-infiltrating cells, among which macrophages are significant components, due to their diversity of functions through which they can influence the immune response against tumor cells. Macrophages present in tumor environment are termed as tumor-associated macrophages (TAMs). They are strongly plastic cells, and depending on the TME stimuli (i.e., cytokines, chemokines), TAMs polarize to antitumoral (M1-like TAMs) or protumoral (M2-like TAMs) phenotype. Both types of TAMs differ in the surface receptors' expression, activation of intracellular signaling pathways, and ability of production and various metabolites release. At the early stage of tumor formation, TAMs are M1-like phenotype, and they are able to eliminate tumor cells, i.e., by reactive oxygen species formation or by presentation of cancer antigens to other effector immune cells. However, during tumor progression, TAMs M2-like phenotype is dominating. They mainly contribute to angiogenesis, stromal remodeling, enhancement of tumor cells migration and invasion, and immunosuppression. This wide variety of TAMs' functions makes them an excellent subject for use in developing antitumor therapies which mainly is based on three strategies: TAMs' elimination, reprograming, or recruitment inhibition.
Collapse
Affiliation(s)
| | - Michal Kielbik
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland.
| |
Collapse
|
29
|
Thangam R, Patel KD, Kang H, Paulmurugan R. Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy-Current Status and Future Perspectives. Vaccines (Basel) 2021; 9:vaccines9080935. [PMID: 34452059 PMCID: PMC8402739 DOI: 10.3390/vaccines9080935] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022] Open
Abstract
Engineering polymeric nanoparticles for their shape, size, surface chemistry, and functionalization using various targeting molecules has shown improved biomedical applications for nanoparticles. Polymeric nanoparticles have created tremendous therapeutic platforms, particularly applications related to chemo- and immunotherapies in cancer. Recently advancements in immunotherapies have broadened this field in immunology and biomedical engineering, where "immunoengineering" creates solutions to target translational science. In this regard, the nanoengineering field has offered the various techniques necessary to manufacture and assemble multifunctional polymeric nanomaterial systems. These include nanoparticles functionalized using antibodies, small molecule ligands, targeted peptides, proteins, and other novel agents that trigger and encourage biological systems to accept the engineered materials as immune enhancers or as vaccines to elevate therapeutic functions. Strategies to engineer polymeric nanoparticles with therapeutic and targeting molecules can provide solutions for developing immune vaccines via maintaining the receptor storage in T- and B cells. Furthermore, cancer immunotherapy using polymeric nanomaterials can serve as a gold standard approach for treating primary and metastasized tumors. The current status of the limited availability of immuno-therapeutic drugs highlights the importance of polymeric nanomaterial platforms to improve the outcomes via delivering anticancer agents at localized sites, thereby enhancing the host immune response in cancer therapy. This review mainly focuses on the potential scientific enhancements and recent developments in cancer immunotherapies by explicitly discussing the role of polymeric nanocarriers as nano-vaccines. We also briefly discuss the role of multifunctional nanomaterials for their therapeutic impacts on translational clinical applications.
Collapse
Affiliation(s)
- Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Correspondence: (R.T.); (R.P.)
| | - Kapil D. Patel
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Correspondence: (R.T.); (R.P.)
| |
Collapse
|
30
|
Mulens-Arias V, Rojas JM, Barber DF. The Use of Iron Oxide Nanoparticles to Reprogram Macrophage Responses and the Immunological Tumor Microenvironment. Front Immunol 2021; 12:693709. [PMID: 34177955 PMCID: PMC8221395 DOI: 10.3389/fimmu.2021.693709] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The synthesis and functionalization of iron oxide nanoparticles (IONPs) is versatile, which has enhanced the interest in studying them as theranostic agents over recent years. As IONPs begin to be used for different biomedical applications, it is important to know how they affect the immune system and its different cell types, especially their interaction with the macrophages that are involved in their clearance. How immune cells respond to therapeutic interventions can condition the systemic and local tissue response, and hence, the final therapeutic outcome. Thus, it is fundamental to understand the effects that IONPs have on the immune response, especially in cancer immunotherapy. The biological effects of IONPs may be the result of intrinsic features of their iron oxide core, inducing reactive oxygen species (ROS) and modulating intracellular redox and iron metabolism. Alternatively, their effects are driven by the nanoparticle coating, for example, through cell membrane receptor engagement. Indeed, exploiting these properties of IONPs could lead to the development of innovative therapies. In this review, after a presentation of the elements that make up the tumor immunological microenvironment, we will review and discuss what is currently known about the immunomodulatory mechanisms triggered by IONPs, mainly focusing on macrophage polarization and reprogramming. Consequently, we will discuss the implications of these findings in the context of plausible therapeutic scenarios for cancer immunotherapy.
Collapse
Affiliation(s)
- Vladimir Mulens-Arias
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - José Manuel Rojas
- Centro de Investigación en Sanidad Animal, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (CISA-INIA)-CSIC, Valdeolmos, Madrid, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| |
Collapse
|
31
|
Meng Y, Zhang D, Sun Y, Dai Z, Zhang T, Yu D, Zhang G, Zheng X. Core-shell FePt-cube@covalent organic polymer nanocomposites: a multifunctional nanocatalytic agent for primary and metastatic tumor treatment. J Mater Chem B 2021; 8:11021-11032. [PMID: 33191409 DOI: 10.1039/d0tb01981j] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metastasis and spread are currently the main factors leading to high mortality of cancer, so developing a synergetic antitumor strategy with high specificity and hypotoxicity is in urgent demand. Based on the design concept of "nanocatalytic medicine", multifunctional nanotherapeutic agent FePt@COP-FA nanocomposites (FPCF NCs) are developed for cancer treatment. Specifically, in the tumor microenvironment (TME), FePt could catalyze intracellular over-expressed H2O2 to generate highly active hydroxyl radicals (˙OH), which could not only induce the apoptosis of tumor cells, but also activate the "ferroptosis" pathway resulting in the lipid peroxide accumulation and ferroptotic cell death. Moreover, owing to the excellent photothermal effect, the FPCF NCs could effectively ablate primary tumors under near-infrared (NIR) laser irradiation and produce numerous tumor-associated antigens in situ. With the assistance of a checkpoint blockade inhibitor, anti-CTLA4 antibody, the body's specific immune response would be initiated to inhibit the growth of metastatic tumors. In particular, such synergistic therapeutics could produce an effective immunological memory effect, which could prevent tumor metastasis and recurrence again. In summary, the FPCF NC is an effective multifunctional antitumor therapeutic agent for nanocatalytic/photothermal/checkpoint blockade combination therapy, which exhibits great potential in nanocatalytic anticancer therapeutic applications.
Collapse
Affiliation(s)
- Yanfei Meng
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P. R. China. and School of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P. R. China
| | - Dongsheng Zhang
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P. R. China.
| | - Yunqiang Sun
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P. R. China. and School of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P. R. China
| | - Zhichao Dai
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P. R. China. and School of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P. R. China
| | - Tiantian Zhang
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P. R. China.
| | - Dexin Yu
- Radiology Departments, Qilu Hospital of Shandong University, Jinan, Shandong 250000, China
| | - Gaorui Zhang
- Radiology Departments, Qilu Hospital of Shandong University, Jinan, Shandong 250000, China
| | - Xiuwen Zheng
- Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, Shandong, P. R. China. and School of Chemistry & Chemical Engineering, Linyi University, Linyi 276000, Shandong, P. R. China
| |
Collapse
|
32
|
Li Q, Li J, Song S, Chen W, Shen X, Li S, Xing D. Nanoparticle-mediated tumor vaccines for personalized therapy: preparing tumor antigens in vivo or ex vivo? J Mater Chem B 2021; 9:2352-2366. [PMID: 33659970 DOI: 10.1039/d0tb02915g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Tumor vaccines, focusing on tailoring individual tumor antigens, have gained much attention in personalized tumor therapy. Recently, breakthroughs have been made in the development of tumor vaccines thanks to the progress in nanotechnology. We will summarize nanoparticle-mediated tumor vaccines for personalized therapy in this review. ROS/heat generating nanoparticles and molecules could induce immunogenic cell death and tumor antigen release in vivo. This strategy often includes chemotherapy, radiotherapy, photodynamic therapy, photothermal therapy, magneto-thermal therapy, etc. On the other hand, ex vivo technologies have been applied for processing of tumor cells/tissues to form effective tumor antigens, in which nanotechnology has shown very good prospects in delivering tumor antigens. In in vivo and ex vivo strategies, nanotechnology also could improve the immune effect through enhancing the uptake by targeting cells, reducing therapeutic drugs/agents, further encapsulating immuno-modulatory molecules or combining with other therapy treatments. Thus, therapeutic vaccines based on nanoparticles have the potential to enhance the immune response and reduce the side effects.
Collapse
Affiliation(s)
- Qian Li
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, 266071, China and Qingdao Cancer Institute, Qingdao, 266071, China
| | - Jia Li
- Qingdao Cancer Institute, Qingdao, 266071, China
| | - Sha Song
- Qingdao Cancer Institute, Qingdao, 266071, China
| | - Wujun Chen
- Qingdao Cancer Institute, Qingdao, 266071, China
| | - Xin Shen
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, 266071, China and CP Pharmaceutical (Qingdao) Co., Ltd, Qingdao, 266426, China.
| | - Suming Li
- Institut Européen des Membranes, IEM-UMR 5635, Univ Montpellier, ENSCM,CNRS, 34095 Montpellier, France.
| | - Dongming Xing
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, 266071, China and Qingdao Cancer Institute, Qingdao, 266071, China and School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
33
|
Dai J, Wu M, Wang Q, Ding S, Dong X, Xue L, Zhu Q, Zhou J, Xia F, Wang S, Hong Y. Red blood cell membrane-camouflaged nanoparticles loaded with AIEgen and Poly(I : C) for enhanced tumoral photodynamic-immunotherapy. Natl Sci Rev 2021; 8:nwab039. [PMID: 34691671 PMCID: PMC8288176 DOI: 10.1093/nsr/nwab039] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/18/2022] Open
Abstract
Red blood cell (RBC)-mimicking nanoparticles (NPs) offer a promising platform for drug delivery because of their prolonged circulation time, reduced immunogenicity and specific targeting ability. Herein, we report the design and preparation of RBC membrane-bound NPs (M@AP), for tumoral photodynamic-immunotherapy. The M@AP is formed by self-assembly of the positively charged aggregation-induced emission luminogen (AIEgen) (named P2-PPh3) and the negatively charged polyinosinic : polycytidylic acid (Poly(I : C)), followed by RBC membrane encapsulation. P2-PPh3 is an AIE-active conjugated polyelectrolyte with additional photosensitizing ability for photodynamic therapy (PDT), while Poly(I : C) serves as an immune-stimulant to stimulate both tumor and immune cells to activate immunity, and thus reduces tumor cell viability. When applied in tumor-bearing mice, the M@AP NPs are enriched in both the tumor region as a result of an enhanced permeability and retention (EPR) effect, and the spleen because of the homing effect of the RBC-mimicking shell. Upon light irradiation, P2-PPh3 promotes strong ROS generation in tumor cells, inducing the release of tumor antigens (TA). The anti-tumor immunity is further enhanced by the presence of Poly(I : C) in M@AP. Thus, this strategy combines the PDT properties of the AIE-active polyelectrolyte and immunotherapy properties of Poly(I : C) to achieve synergistic activation of the immune system for anti-tumor activity, providing a novel strategy for tumor treatment.
Collapse
Affiliation(s)
- Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Quan Wang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Siyang Ding
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Xiaoqi Dong
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Qingqing Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Jian Zhou
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Fan Xia
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Yuning Hong
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| |
Collapse
|
34
|
Seaberg J, Montazerian H, Hossen MN, Bhattacharya R, Khademhosseini A, Mukherjee P. Hybrid Nanosystems for Biomedical Applications. ACS NANO 2021; 15:2099-2142. [PMID: 33497197 PMCID: PMC9521743 DOI: 10.1021/acsnano.0c09382] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Inorganic/organic hybrid nanosystems have been increasingly developed for their versatility and efficacy at overcoming obstacles not readily surmounted by nonhybridized counterparts. Currently, hybrid nanosystems are implemented for gene therapy, drug delivery, and phototherapy in addition to tissue regeneration, vaccines, antibacterials, biomolecule detection, imaging probes, and theranostics. Though diverse, these nanosystems can be classified according to foundational inorganic/organic components, accessory moieties, and architecture of hybridization. Within this Review, we begin by providing a historical context for the development of biomedical hybrid nanosystems before describing the properties, synthesis, and characterization of their component building blocks. Afterward, we introduce the architectures of hybridization and highlight recent biomedical nanosystem developments by area of application, emphasizing hybrids of distinctive utility and innovation. Finally, we draw attention to ongoing clinical trials before recapping our discussion of hybrid nanosystems and providing a perspective on the future of the field.
Collapse
Affiliation(s)
- Joshua Seaberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
| | - Hossein Montazerian
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Md Nazir Hossen
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
35
|
Zhang H, Tang WL, Kheirolomoom A, Fite BZ, Wu B, Lau K, Baikoghli M, Raie MN, Tumbale SK, Foiret J, Ingham ES, Mahakian LM, Tam SM, Cheng RH, Borowsky AD, Ferrara KW. Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J Control Release 2021; 330:1080-1094. [PMID: 33189786 PMCID: PMC7906914 DOI: 10.1016/j.jconrel.2020.11.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/01/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022]
Abstract
Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 μg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Wei-Lun Tang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Azadeh Kheirolomoom
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Kenneth Lau
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Marina Nura Raie
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Spencer K Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | | | - Katherine W Ferrara
- Molecular Imaging Program, Department of Radiology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
36
|
Cheng H, Tsao H, Chiang C, Chen S. Advances in Magnetic Nanoparticle-Mediated Cancer Immune-Theranostics. Adv Healthc Mater 2021; 10:e2001451. [PMID: 33135398 DOI: 10.1002/adhm.202001451] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/12/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is a cutting-edge strategy that eliminates cancer cells by amplifying the host's immune system. However, the low response rate and risks of inducing systemic toxicity have raised uncertainty in the treatment. Magnetic nanoparticles (MNPs) as a versatile theranostic tool can be used to target delivery of multiple immunotherapeutics and monitor cell/tissue responses. These capabilities enable the real-time characterization of the factors that contribute to immunoactivity so that future treatments can be optimized. The magnetic properties of MNPs further allow the implementation of magnetic navigation and magnetic hyperthermia for boosting the efficacy of immunotherapy. The multimodal approach opens an avenue to induce robust immune responses, minimize safety issues, and monitor immune activities simultaneously. Thus, the object of this review is to provide an overview of the burgeoning fields and to highlight novel technologies for next-generation immunotherapy. The review further correlates the properties of MNPs with the latest treatment strategies to explore the crosstalk between magnetic nanomaterials and the immune system. This comprehensive review of MNP-derived immunotherapy covers the obstacles and opportunities for future development and clinical translation.
Collapse
Affiliation(s)
- Hung‐Wei Cheng
- Department of Materials Science and Engineering National Chiao Tung University Hsinchu 30010 Taiwan
| | - Hsin‐Yi Tsao
- Department of Materials Science and Engineering National Chiao Tung University Hsinchu 30010 Taiwan
| | - Chih‐Sheng Chiang
- Cell Therapy Center China Medical University Hospital Taichung 40421 Taiwan
| | - San‐Yuan Chen
- Department of Materials Science and Engineering National Chiao Tung University Hsinchu 30010 Taiwan
- Frontier Research Centre on Fundamental and Applied Sciences of Matters National Tsing Hua University Hsinchu 30013 Taiwan
- School of Dentistry College of Dental Medicine Kaohsiung Medical University Kaohsiung 807378 Taiwan
- Graduate Institute of Biomedical Science China Medical University Taichung 40421 Taiwan
| |
Collapse
|
37
|
Thakur N, Thakur S, Chatterjee S, Das J, Sil PC. Nanoparticles as Smart Carriers for Enhanced Cancer Immunotherapy. Front Chem 2020; 8:597806. [PMID: 33409265 PMCID: PMC7779678 DOI: 10.3389/fchem.2020.597806] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy for the treatment of many forms of cancer by stimulating body's own immune system. This therapy not only eradicates tumor cells by inducing strong anti-tumor immune response but also prevent their recurrence. The clinical cancer immunotherapy faces some insurmountable challenges including high immune-mediated toxicity, lack of effective and targeted delivery of cancer antigens to immune cells and off-target side effects. However, nanotechnology offers some solutions to overcome those limitations, and thus can potentiate the efficacy of immunotherapy. This review focuses on the advancement of nanoparticle-mediated delivery of immunostimulating agents for efficient cancer immunotherapy. Here we have outlined the use of the immunostimulatory nanoparticles as a smart carrier for effective delivery of cancer antigens and adjuvants, type of interactions between nanoparticles and the antigen/adjuvant as well as the factors controlling the interaction between nanoparticles and the receptors on antigen presenting cells. Besides, the role of nanoparticles in targeting/activating immune cells and modulating the immunosuppressive tumor microenvironment has also been discussed extensively. Finally, we have summarized some theranostic applications of the immunomodulatory nanomaterials in treating cancers based on the earlier published reports.
Collapse
Affiliation(s)
- Neelam Thakur
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- School of Advanced Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, India
| | - Saloni Thakur
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India
| | | | - Joydeep Das
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- School of Advanced Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, India
| | - Parames C. Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
38
|
Ahmad MZ, Ahmad J, Haque A, Alasmary MY, Abdel-Wahab BA, Akhter S. Emerging advances in synthetic cancer nano-vaccines: opportunities and challenges. Expert Rev Vaccines 2020; 19:1053-1071. [DOI: 10.1080/14760584.2020.1858058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Anzarul Haque
- Department of Pharmacognosy, Prince Sattam Bin Abdulaziz University College of Pharmacy, Alkharj Al-Kharj, Kingdom of Saudi Arabia
| | - Mohammed Yahia Alasmary
- Department of Internal Medicine, College of Medicine, Najran University Hospital, Najran, Kingdom of Saudi Arabia
| | - Basel A. Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
- Department of Pharmacology, College of Medicine Assiut University, Assiut, Egypt
| | - Sohail Akhter
- Center for Molecular Biophysics (CBM), CNRS UPR4301; LE STUDIUM Loire Valley Institute for Advanced Studies, Orleans, France
| |
Collapse
|
39
|
Cheng X, Li H, Ge X, Chen L, Liu Y, Mao W, Zhao B, Yuan WE. Tumor-Microenvironment- Responsive Size-Shrinkable Drug-Delivery Nanosystems for Deepened Penetration Into Tumors. Front Mol Biosci 2020; 7:576420. [PMID: 33330618 PMCID: PMC7729065 DOI: 10.3389/fmolb.2020.576420] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/13/2020] [Indexed: 01/04/2023] Open
Abstract
Over the years, the manipulation and clinical application of drug-delivery nanosystems for cancer diseases have attracted a rapid growth of academic research interests, and some nanodrugs have been approved for clinic application. Although encouraging achievements have been made, the potency of nanomedicines in cancer treatment is far from satisfaction, and one significant reason is the inefficient penetration of nanoparticles into solid tumors. Particle size is one of the most significant features that influence diffusion ability of the drug-delivery system in tumors. Size-shrinkable drug-delivery nanosystems possess a size-switchable property that can achieve passive targeting via the enhanced permeability and retention (EPR) effect and transform into ultrasmall particles in tumors for deep penetration into tumors. The tumor microenvironment is characterized by acidic pH, hypoxia, upregulated levels of enzymes, and a redox environment. In this review, we summarize and analyze the current research progresses and challenges in tumor microenvironment responsive size-shrinkable drug-delivery nanosystems. We further expect to present some meaningful proposals and enlightenments on promoting deep penetration into tumors of nanoparticles.
Collapse
Affiliation(s)
- Xiaoliang Cheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Houli Li
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuemei Ge
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, China
| | - Lijuan Chen
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, China
| | - Yao Liu
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, China
| | - Wenwei Mao
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Zhao
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-En Yuan
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
40
|
Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B 2020; 10:2110-2124. [PMID: 33304781 PMCID: PMC7714990 DOI: 10.1016/j.apsb.2020.05.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
The complex tumor microenvironment is a most important factor in cancer development. The biological microenvironment is composed of a variety of barriers including the extracellular matrix and associated cells such as endothelia cells, pericytes, and cancer-associated fibroblasts. Different strategies can be utilized to enhance nanoparticle-based drug delivery and distribution into tumor tissues addressing the extracellular matrix or cellular components. In addition to the biological microenvironment, the immunological conditions around the tumor tissue can be very complicated and cancer cells have various ways of evading immune surveillance. Nanoparticle drug delivery systems can enhance cancer immunotherapy by tuning the immunological response and memory of various immune cells such as T cells, B cells, macrophages, and dendritic cells. In this review, the main components in the tumor biological and immunological environment are discussed. The focus is on recent advances in nanoparticle-based drug delivery systems towards targets within the tumor microenvironment to improve cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| | - Diane J. Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
41
|
Ruiz-de-Angulo A, Bilbao-Asensio M, Cronin J, Evans SJ, Clift MJ, Llop J, Feiner IV, Beadman R, Bascarán KZ, Mareque-Rivas JC. Chemically Programmed Vaccines: Iron Catalysis in Nanoparticles Enhances Combination Immunotherapy and Immunotherapy-Promoted Tumor Ferroptosis. iScience 2020; 23:101499. [PMID: 32919370 PMCID: PMC7490994 DOI: 10.1016/j.isci.2020.101499] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/24/2020] [Accepted: 08/21/2020] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy has yielded impressive results, but only for a minority of patients with cancer. Therefore, new approaches that potentiate immunotherapy are a pressing medical need. Ferroptosis is a newly described type of programmed cell death driven by iron-dependent phospholipid peroxidation via Fenton chemistry. Here, we developed iron oxide-loaded nanovaccines (IONVs), which, chemically programmed to integrate iron catalysis, drug delivery, and tracking exploiting the characteristics of the tumor microenvironment (TME), improves immunotherapy and activation of ferroptosis. The IONVs trigger danger signals and use molecular disassembly and reversible covalent bonds for targeted antigen delivery and improved immunostimulatory capacity and catalytic iron for targeting tumor cell ferroptosis. IONV- and antibody-mediated TME modulation interfaced with imaging was important toward achieving complete eradication of aggressive and established tumors, eliciting long-lived protective antitumor immunity with no toxicities. This work establishes the feasibility of using nanoparticle iron catalytic activity as a versatile and effective feature for enhancing immunotherapy.
Collapse
Affiliation(s)
- Ane Ruiz-de-Angulo
- Chemical Immunology Laboratory, CIC BioGUNE, Building 801A, Derio 48160, Spain
| | - Marc Bilbao-Asensio
- Department of Chemistry and Centre for NanoHealth, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - James Cronin
- Swansea University Medical School, Institute of Life Science, Singleton Park, Swansea SA2 8PP, UK
| | - Stephen J. Evans
- Swansea University Medical School, Institute of Life Science, Singleton Park, Swansea SA2 8PP, UK
| | - Martin J.D. Clift
- Swansea University Medical School, Institute of Life Science, Singleton Park, Swansea SA2 8PP, UK
| | - Jordi Llop
- Radiochemistry and Nuclear Imaging Laboratory, CIC BiomaGUNE, Paseo Miramón 182, San Sebastián 20014, Spain
| | - Irene V.J. Feiner
- Radiochemistry and Nuclear Imaging Laboratory, CIC BiomaGUNE, Paseo Miramón 182, San Sebastián 20014, Spain
| | - Rhiannon Beadman
- Swansea University Medical School, Institute of Life Science, Singleton Park, Swansea SA2 8PP, UK
| | - Kepa Zamacola Bascarán
- Radiochemistry and Nuclear Imaging Laboratory, CIC BiomaGUNE, Paseo Miramón 182, San Sebastián 20014, Spain
| | - Juan C. Mareque-Rivas
- Department of Chemistry and Centre for NanoHealth, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| |
Collapse
|
42
|
Seth A, Derami HG, Gupta P, Wang Z, Rathi P, Gupta R, Cao T, Morrissey JJ, Singamaneni S. Polydopamine-Mesoporous Silica Core-Shell Nanoparticles for Combined Photothermal Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:42499-42510. [PMID: 32838525 PMCID: PMC7942218 DOI: 10.1021/acsami.0c10781] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cancer immunotherapy involves a cascade of events that ultimately leads to cytotoxic immune cells effectively identifying and destroying cancer cells. Responsive nanomaterials, which enable spatiotemporal orchestration of various immunological events for mounting a highly potent and long-lasting antitumor immune response, are an attractive platform to overcome challenges associated with existing cancer immunotherapies. Here, we report a multifunctional near-infrared (NIR)-responsive core-shell nanoparticle, which enables (i) photothermal ablation of cancer cells for generating tumor-associated antigen (TAA) and (ii) triggered release of an immunomodulatory drug (gardiquimod) for starting a series of immunological events. The core of these nanostructures is composed of a polydopamine nanoparticle, which serves as a photothermal agent, and the shell is made of mesoporous silica, which serves as a drug carrier. We employed a phase-change material as a gatekeeper to achieve concurrent release of both TAA and adjuvant, thus efficiently activating the antigen-presenting cells. Photothermal immunotherapy enabled by these nanostructures resulted in regression of primary tumor and significantly improved inhibition of secondary tumor in a mouse melanoma model. These biocompatible, biodegradable, and NIR-responsive core-shell nanostructures simultaneously deliver payload and cause photothermal ablation of the cancer cells. Our results demonstrate potential of responsive nanomaterials in generating highly synergistic photothermal immunotherapeutic response.
Collapse
Affiliation(s)
- Anushree Seth
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hamed Gholami Derami
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Prashant Gupta
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Zheyu Wang
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Priya Rathi
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Rohit Gupta
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Thao Cao
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jeremiah J. Morrissey
- Department of Anesthesiology, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
43
|
Yang F, Shi K, Jia YP, Hao Y, Peng JR, Qian ZY. Advanced biomaterials for cancer immunotherapy. Acta Pharmacol Sin 2020; 41:911-927. [PMID: 32123302 PMCID: PMC7468530 DOI: 10.1038/s41401-020-0372-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/27/2020] [Indexed: 02/05/2023] Open
Abstract
Immunotherapy, as a powerful strategy for cancer treatment, has achieved tremendous efficacy in clinical trials. Despite these advancements, there is much to do in terms of enhancing therapeutic benefits and decreasing the side effects of cancer immunotherapy. Advanced nanobiomaterials, including liposomes, polymers, and silica, play a vital role in the codelivery of drugs and immunomodulators. These nanobiomaterial-based delivery systems could effectively promote antitumor immune responses and simultaneously reduce toxic adverse effects. Furthermore, nanobiomaterials may also combine with each other or with traditional drugs via different mechanisms, thus giving rise to more accurate and efficient tumor treatment. Here, an overview of the latest advancement in these nanobiomaterials used for cancer immunotherapy is given, describing outstanding systems, including lipid-based nanoparticles, polymer-based scaffolds or micelles, inorganic nanosystems, and others.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Yan-Peng Jia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Ying Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Jin-Rong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zhi-Yong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
44
|
Wang L, He Y, He T, Liu G, Lin C, Li K, Lu L, Cai K. Lymph node-targeted immune-activation mediated by imiquimod-loaded mesoporous polydopamine based-nanocarriers. Biomaterials 2020; 255:120208. [PMID: 32569862 DOI: 10.1016/j.biomaterials.2020.120208] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022]
Abstract
Toll-like receptor (TLR) agonists are the potent stimulants of innate immune system and hold promises as an adjuvant for anticancer immunotherapy. Unfortunately, most of them are limited by a prompt dissemination, and thus caused "wasted inflammation". Hence, how to restrict their action radius into lymphoid tissues is of great relevance to enhance their efficacy and concomitantly alleviates the side effects. Here, imiquimod (R837), a TLR 7 agonist, was loaded into mesoporous polydopamine (MPDA) nanocarriers with high efficiency. Moreover, its surface was modified by polyvinyl pyrrolidone (PVP) to enhance their lymphatic drainage ability. These nano-adjuvants have obvious advantages in promoting dendritic cell (DC) maturation in comparison to free R837. Moreover, their transportation and retention ability in proximal lymph nodes (LNs) were also confirmed, by which lymphatic drug exposure can be maximized to a great extent. Consequently, effective DC activation and CD8+ T cell responses were observed as expected by R837 released in draining LNs. This effect was further enhanced by the presence of endogenous tumor antigens from apoptosis debris induced by MPDA-based photothermal effect, and thus led to the growth inhibition of subcutaneous B16 melanomas. The results demonstrated the great potency against melanoma of the designed PVP-MPDA@R837 nano-adjuvants by combining photothermal conversion property of MPDA with lymphatic-focused immune-activation.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Ye He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tingting He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Genhua Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chuanchua Lin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Ke Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Lu Lu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
45
|
Jin JO, Kim G, Hwang J, Han KH, Kwak M, Lee PCW. Nucleic acid nanotechnology for cancer treatment. Biochim Biophys Acta Rev Cancer 2020; 1874:188377. [PMID: 32418899 DOI: 10.1016/j.bbcan.2020.188377] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022]
Abstract
Cancer is one of the most prevalent potentially lethal diseases. With the increase in the number of investigations into the uses of nanotechnology, many nucleic acid (NA)-based nanostructures such as small interfering RNA, microRNA, aptamers, and immune adjuvant NA have been applied to treat cancer. Here, we discuss studies on the applications of NA in cancer treatment, recent research trends, and the limitations and prospects of specific NA-mediated gene therapy and immunotherapy for cancer treatment. The NA structures used for cancer therapy consist only of NA or hybrids comprising organic or inorganic substances integrated with functional NA. We also discuss delivery vehicles for therapeutic NA and anti-cancer agents, and recent trends in NA-based gene therapy and immunotherapy against cancer.
Collapse
Affiliation(s)
- Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China; Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea.
| | - Gyurin Kim
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - Juyoung Hwang
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| | - Kyung Ho Han
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea; DWI-Leibniz Institute for Interactive Materials, Aachen 52056, Germany.
| | - Peter C W Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea.
| |
Collapse
|
46
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
47
|
Santos-Sierra S. Developments in anticancer vaccination: budding new adjuvants. Biol Chem 2020; 401:435-446. [DOI: 10.1515/hsz-2019-0383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/21/2019] [Indexed: 12/17/2022]
Abstract
AbstractThe immune system has a limited capacity to recognize and fight cells that become cancerous and in cancer patients, the immune system has to seek the right balance between cancer rejection and host-immunosupression. The tumor milieu builds a protective shell and tumor cells rapidly accumulate mutations that promote antigen variability and immune-escape. Therapeutic vaccination of cancer is a promising strategy the success of which depends on a powerful activation of the cells of the adaptive immune system specific for tumor-cell detection and killing (e.g. CD4+and CD8+T-cells). In the last decades, the search for novel adjuvants that enhance dendritic cell (DC) function and their ability to prime T-cells has flourished and some Toll-like receptor (TLR) agonists have long been known to be valid immune adjuvants. The implementation of TLR-synthetic agonists in clinical studies of cancer vaccination is replacing the initial use of microbial-derived products with some encouraging results. The purpose of this review is to summarize the latest discoveries of TLR-synthetic agonists with adjuvant potential in anti-cancer vaccination.
Collapse
Affiliation(s)
- Sandra Santos-Sierra
- Section of Biochemical Pharmacology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| |
Collapse
|
48
|
Mukherjee S, Liang L, Veiseh O. Recent Advancements of Magnetic Nanomaterials in Cancer Therapy. Pharmaceutics 2020; 12:pharmaceutics12020147. [PMID: 32053995 PMCID: PMC7076668 DOI: 10.3390/pharmaceutics12020147] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/05/2020] [Accepted: 02/08/2020] [Indexed: 12/16/2022] Open
Abstract
Magnetic nanomaterials belong to a class of highly-functionalizable tools for cancer therapy owing to their intrinsic magnetic properties and multifunctional design that provides a multimodal theranostics platform for cancer diagnosis, monitoring, and therapy. In this review article, we have provided an overview of the various applications of magnetic nanomaterials and recent advances in the development of these nanomaterials as cancer therapeutics. Moreover, the cancer targeting, potential toxicity, and degradability of these nanomaterials has been briefly addressed. Finally, the challenges for clinical translation and the future scope of magnetic nanoparticles in cancer therapy are discussed.
Collapse
|
49
|
Application of molecular imaging technology in tumor immunotherapy. Cell Immunol 2020; 348:104039. [DOI: 10.1016/j.cellimm.2020.104039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/21/2019] [Accepted: 01/07/2020] [Indexed: 02/08/2023]
|
50
|
DeLong RK, Dean J, Glaspell G, Jaberi-Douraki M, Ghosh K, Davis D, Monteiro-Riviere N, Chandran P, Nguyen T, Aryal S, Middaugh CR, Chan Park S, Choi SO, Ramani M. Amino/Amido Conjugates Form to Nanoscale Cobalt Physiometacomposite (PMC) Materials Functionally Delivering Nucleic Acid Therapeutic to Nucleus Enhancing Anticancer Activity via Ras-Targeted Protein Interference. ACS APPLIED BIO MATERIALS 2020; 3:175-179. [DOI: 10.1021/acsabm.9b00798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Robert K. DeLong
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - John Dean
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| | - Garry Glaspell
- US Army Corps of Engineers Engineer Research & Development Center, Alexandria, Virginia 22315, United States
| | - Majid Jaberi-Douraki
- Institute for Computational Comparative Medicine, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - Kartik Ghosh
- Physics and Materials Science, Missouri State University, Springfield, Missouri 65897, United States
| | - Daniel Davis
- Animal Modeling Core, University of Missouri, Columbia, Missouri 65201, United States
| | - Nancy Monteiro-Riviere
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - Parwathy Chandran
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - Tuyen Nguyen
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - Santosh Aryal
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - C. Russell Middaugh
- Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 67047, United States
| | - Seok Chan Park
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - Seong-O Choi
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| | - Meghana Ramani
- Nanotechnology Innovation Center, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66502, United States
| |
Collapse
|