1
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Lani R, Thariq IM, Suhaimi NS, Hassandarvish P, Abu Bakar S. From defense to offense: Modulating toll-like receptors to combat arbovirus infections. Hum Vaccin Immunother 2024; 20:2306675. [PMID: 38263674 PMCID: PMC11657068 DOI: 10.1080/21645515.2024.2306675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/08/2024] [Accepted: 01/14/2024] [Indexed: 01/25/2024] Open
Abstract
Arboviruses are a significant threat to global public health, with outbreaks occurring worldwide. Toll-like receptors (TLRs) play a crucial role in the innate immune response against these viruses by recognizing pathogen-associated molecular patterns and initiating an inflammatory response. Significantly, TLRs commonly implicated in the immune response against viral infections include TLR2, TLR4, TLR6, TLR3, TLR7, and TLR8; limiting or allowing them to replicate and spread within the host. Modulating TLRs has emerged as a promising approach to combat arbovirus infections. This review summarizes recent advances in TLR modulation as a therapeutic target in arbovirus infections. Studies have shown that the activation of TLRs can enhance the immune response against arbovirus infections, leading to increased viral clearance and protection against disease. Conversely, inhibition of TLRs can reduce the excessive inflammation and tissue damage associated with arbovirus infection. Modulating TLRs represents a potential therapeutic strategy to combat arbovirus infections.
Collapse
Affiliation(s)
- Rafidah Lani
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ilya Maisarah Thariq
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Nuramira Syazreen Suhaimi
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Pouya Hassandarvish
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Sazaly Abu Bakar
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
Malik S, Sureka N, Ahuja S, Aden D, Zaheer S, Zaheer S. Tumor-associated macrophages: A sentinel of innate immune system in tumor microenvironment gone haywire. Cell Biol Int 2024; 48:1406-1449. [PMID: 39054741 DOI: 10.1002/cbin.12226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
The tumor microenvironment (TME) is a critical determinant in the initiation, progression, and treatment outcomes of various cancers. Comprising of cancer-associated fibroblasts (CAF), immune cells, blood vessels, and signaling molecules, the TME is often likened to the soil supporting the seed (tumor). Among its constituents, tumor-associated macrophages (TAMs) play a pivotal role, exhibiting a dual nature as both promoters and inhibitors of tumor growth. This review explores the intricate relationship between TAMs and the TME, emphasizing their diverse functions, from phagocytosis and tissue repair to modulating immune responses. The plasticity of TAMs is highlighted, showcasing their ability to adopt either protumorigenic or anti-tumorigenic phenotypes based on environmental cues. In the context of cancer, TAMs' pro-tumorigenic activities include promoting angiogenesis, inhibiting immune responses, and fostering metastasis. The manuscript delves into therapeutic strategies targeting TAMs, emphasizing the challenges faced in depleting or inhibiting TAMs due to their multifaceted roles. The focus shifts towards reprogramming TAMs to an anti-tumorigenic M1-like phenotype, exploring interventions such as interferons, immune checkpoint inhibitors, and small molecule modulators. Noteworthy advancements include the use of CSF1R inhibitors, CD40 agonists, and CD47 blockade, demonstrating promising results in preclinical and clinical settings. A significant section is dedicated to Chimeric Antigen Receptor (CAR) technology in macrophages (CAR-M cells). While CAR-T cells have shown success in hematological malignancies, their efficacy in solid tumors has been limited. CAR-M cells, engineered to infiltrate solid tumors, are presented as a potential breakthrough, with a focus on their development, challenges, and promising outcomes. The manuscript concludes with the exploration of third-generation CAR-M technology, offering insight into in-vivo reprogramming and nonviral vector approaches. In conclusion, understanding the complex and dynamic role of TAMs in cancer is crucial for developing effective therapeutic strategies. While early-stage TAM-targeted therapies show promise, further extensive research and larger clinical trials are warranted to optimize their targeting and improve overall cancer treatment outcomes.
Collapse
Affiliation(s)
- Shaivy Malik
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, Jamia Hamdard, New Delhi, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| |
Collapse
|
4
|
Zhang WY, Zheng XL, Coghi PS, Chen JH, Dong BJ, Fan XX. Revolutionizing adjuvant development: harnessing AI for next-generation cancer vaccines. Front Immunol 2024; 15:1438030. [PMID: 39206192 PMCID: PMC11349682 DOI: 10.3389/fimmu.2024.1438030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
With the COVID-19 pandemic, the importance of vaccines has been widely recognized and has led to increased research and development efforts. Vaccines also play a crucial role in cancer treatment by activating the immune system to target and destroy cancer cells. However, enhancing the efficacy of cancer vaccines remains a challenge. Adjuvants, which enhance the immune response to antigens and improve vaccine effectiveness, have faced limitations in recent years, resulting in few novel adjuvants being identified. The advancement of artificial intelligence (AI) technology in drug development has provided a foundation for adjuvant screening and application, leading to a diversification of adjuvants. This article reviews the significant role of tumor vaccines in basic research and clinical treatment and explores the use of AI technology to screen novel adjuvants from databases. The findings of this review offer valuable insights for the development of new adjuvants for next-generation vaccines.
Collapse
Affiliation(s)
- Wan-Ying Zhang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Xiao-Li Zheng
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Paolo Saul Coghi
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jun-Hui Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bing-Jun Dong
- Gynecology Department, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Xing-Xing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
5
|
Naskar S, Harsukhbhai Chandpa H, Agarwal S, Meena J. Super epitope dengue vaccine instigated serotype independent immune protection in-silico. Vaccine 2024; 42:3857-3873. [PMID: 38616437 DOI: 10.1016/j.vaccine.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 06/14/2024]
Abstract
Dengue becomes the most common life-threatening infectious arbovirus disease globally, with prevalence in the tropical and subtropical areas. The major clinical features include dengue haemorrhagic fever (DHF) and dengue shock syndrome (DSS), a condition of hypovolemic shock. Four different serotypes of the dengue virus, known as dengue virus serotype (DENV)- 1, 2, 3 and 4 can infect humans. Only one vaccine is available in the market, named Dengvaxia by Sanofi Pasteur, but there is no desired outcome of this treatment due the antibody dependent enhancement (ADE) of the multiple dengue serotypes. As of now, there is no cure against dengue disease. Our goal in this work was to create a subunit vaccine based on several epitopes that would be effective against every serotype of the dengue virus. Here, computational methods like- immunoinformatics and bioinformatics were implemented to find out possible dominant epitopes. A total of 21 epitopes were chosen using various in-silico techniques from the expected 133 major histocompatibility complex (MHC)- I and major histocompatibility complex (MHC)- II epitopes, along with 95 B-cell epitopes which were greatly conserved. Immune stimulant, non-allergenic and non-toxic immunodominant epitopes (super epitopes) with a suitable adjuvant (Heparin-Binding Hemagglutinin Adhesin, HBHA) were used to construct the vaccine. Following the physicochemical analysis, vaccine construct was docked with Toll-like receptors (TLRs) to predict the immune stimulation. Consequently, the optimal docked complex that demonstrated the least amount of ligand-receptor complex deformability was used to conduct the molecular dynamics analysis. By following the codon optimization, the final vaccine molecule was administered into an expressing vector to perform in-silico cloning. The robust immune responses were generated in the in-silico immune simulation analysis. Hence, this study provides a hope to control the dengue infections. For validation of the immune outcomes, in-vitro as well as in-vivo investigations are essential.
Collapse
Affiliation(s)
- Shovan Naskar
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Hitesh Harsukhbhai Chandpa
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Shalini Agarwal
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Jairam Meena
- ImmunoEngineering and Therapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India.
| |
Collapse
|
6
|
Cui Y, Ho M, Hu Y, Shi Y. Vaccine adjuvants: current status, research and development, licensing, and future opportunities. J Mater Chem B 2024; 12:4118-4137. [PMID: 38591323 PMCID: PMC11180427 DOI: 10.1039/d3tb02861e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Vaccines represent one of the most significant inventions in human history and have revolutionized global health. Generally, a vaccine functions by triggering the innate immune response and stimulating antigen-presenting cells, leading to a defensive adaptive immune response against a specific pathogen's antigen. As a key element, adjuvants are chemical materials often employed as additives to increase a vaccine's efficacy and immunogenicity. For over 90 years, adjuvants have been essential components in many human vaccines, improving their efficacy by enhancing, modulating, and prolonging the immune response. Here, we provide a timely and comprehensive review of the historical development and the current status of adjuvants, covering their classification, mechanisms of action, and roles in different vaccines. Additionally, we perform systematic analysis of the current licensing processes and highlights notable examples from clinical trials involving vaccine adjuvants. Looking ahead, we anticipate future trends in the field, including the development of new adjuvant formulations, the creation of innovative adjuvants, and their integration into the broader scope of systems vaccinology and vaccine delivery. The article posits that a deeper understanding of biochemistry, materials science, and vaccine immunology is crucial for advancing vaccine technology. Such advancements are expected to lead to the future development of more effective vaccines, capable of combating emerging infectious diseases and enhancing public health.
Collapse
Affiliation(s)
- Ying Cui
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Megan Ho
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Yongjie Hu
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
7
|
Dobrovolskienė N, Balevičius R, Mlynska A, Žilionytė K, Aleksander Krasko J, Strioga M, Lieknina I, Pjanova D, Pašukonienė V. Immunomodulatory properties of bacteriophage derived dsRNA of different size and their use as anticancer vaccine adjuvants. Vaccine 2024; 42:512-521. [PMID: 38184395 DOI: 10.1016/j.vaccine.2023.12.071] [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: 10/04/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024]
Abstract
Dendritic cell (DC) based immunotherapy is one of the strategies to combat cancer invoking a patient's immune system. This form of anticancer immunotherapy employs adjuvants to enhance the immune response, triggering mechanisms of innate immunity and thus increase immunotherapeutic efficiency. A conventional adjuvant for DCs maturation during production of anticancer vaccines is bacterial LPS. Nevertheless, synthetic dsRNAs were also shown to stimulate different receptors on innate immune cells and to activate immune responses through induction of cytokines via toll-like receptors. In our study we investigated the potential of Larifan as dsRNA of natural origin to stimulate maturation of DCs with proinflammatory (possible antitumoral) activity and to compare these immunostimulatory properties between Larifan's fractions with different molecular lengths. To explore the suitability of this product for therapy, it is necessary to study the properties of its different fractions and compare them to standard adjuvants. We investigated the effect of Larifan's fractions on immune system stimulation in vivo by monitoring the survival time of tumor-bearing mice. Murine DCs produced in vitro using Larifan and its fractions together with tumor antigens during production were also characterized. All Larifan fractions resulted in inducing high expression of immunogenic markers CD40, CD80, CD86, CCR7, MHC II and lower secretion of the immunosuppressive cytokine IL-10, compared to the maturation with LPS in mDCs. The lowest expression of tolerogenic gene Ido1 and highest expression of the immunogenic genes Clec7a, Tnf, Icosl, Il12rb2, Cd209a were characteristic to the unfractionated dsRNA and short fraction FR15. In the mouse model the best overall survival rate was observed in mice treated with medium-length FR9 and FR15. We can state that both Larifan and its fractions were superior to LPS as vaccine adjuvants in stimulating phenotype and functional activity of mature DCs. DCs maturation using these factors induces a valuable anticancer immune response.
Collapse
Affiliation(s)
- Neringa Dobrovolskienė
- Laboratory of Immunology, National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania.
| | - Ramojus Balevičius
- Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania.
| | - Agata Mlynska
- Laboratory of Immunology, National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania; Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Saulėtekio al. 11, LT-10223 Vilnius, Lithuania.
| | - Karolina Žilionytė
- Laboratory of Immunology, National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania.
| | - Jan Aleksander Krasko
- Laboratory of Immunology, National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania; Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Saulėtekio al. 11, LT-10223 Vilnius, Lithuania.
| | - Marius Strioga
- Laboratory of Immunology, National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania
| | - Ilva Lieknina
- Latvian Biomedical Research and Study Centre, Ratsupites Street 1, Riga LV-1067, Latvia.
| | - Dace Pjanova
- Latvian Biomedical Research and Study Centre, Ratsupites Street 1, Riga LV-1067, Latvia; Riga Stradins University, Ratsupites street 5., Riga LV-1067, Latvia.
| | - Vita Pašukonienė
- Laboratory of Immunology, National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania; Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Saulėtekio al. 11, LT-10223 Vilnius, Lithuania.
| |
Collapse
|
8
|
Filippou C, Themistocleous SC, Marangos G, Panayiotou Y, Fyrilla M, Kousparou CA, Pana ZD, Tsioutis C, Johnson EO, Yiallouris A. Microbial Therapy and Breast Cancer Management: Exploring Mechanisms, Clinical Efficacy, and Integration within the One Health Approach. Int J Mol Sci 2024; 25:1110. [PMID: 38256183 PMCID: PMC10816061 DOI: 10.3390/ijms25021110] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
This comprehensive review elucidates the profound relationship between the human microbiome and breast cancer management. Recent findings highlight the significance of microbial alterations in tissue, such as the gut and the breast, and their role in influencing the breast cancer risk, development, progression, and treatment outcomes. We delve into how the gut microbiome can modulate systemic inflammatory responses and estrogen levels, thereby impacting cancer initiation and therapeutic drug efficacy. Furthermore, we explore the unique microbial diversity within breast tissue, indicating potential imbalances brought about by cancer and highlighting specific microbes as promising therapeutic targets. Emphasizing a holistic One Health approach, this review underscores the importance of integrating insights from human, animal, and environmental health to gain a deeper understanding of the complex microbe-cancer interplay. As the field advances, the strategic manipulation of the microbiome and its metabolites presents innovative prospects for the enhancement of cancer diagnostics and therapeutics. However, rigorous clinical trials remain essential to confirm the potential of microbiota-based interventions in breast cancer management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Andreas Yiallouris
- School of Medicine, European University Cyprus, 6 Diogenis Str., 2404 Engomi, P.O. Box 22006, Nicosia 1516, Cyprus
| |
Collapse
|
9
|
Gurunathan S, Thangaraj P, Wang L, Cao Q, Kim JH. Nanovaccines: An effective therapeutic approach for cancer therapy. Biomed Pharmacother 2024; 170:115992. [PMID: 38070247 DOI: 10.1016/j.biopha.2023.115992] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Cancer vaccines hold considerable promise for the immunotherapy of solid tumors. Nanomedicine offers several strategies for enhancing vaccine effectiveness. In particular, molecular or (sub) cellular vaccines can be delivered to the target lymphoid tissues and cells by nanocarriers and nanoplatforms to increase the potency and durability of antitumor immunity and minimize negative side effects. Nanovaccines use nanoparticles (NPs) as carriers and/or adjuvants, offering the advantages of optimal nanoscale size, high stability, ample antigen loading, high immunogenicity, tunable antigen presentation, increased retention in lymph nodes, and immunity promotion. To induce antitumor immunity, cancer vaccines rely on tumor antigens, which are administered in the form of entire cells, peptides, nucleic acids, extracellular vesicles (EVs), or cell membrane-encapsulated NPs. Ideal cancer vaccines stimulate both humoral and cellular immunity while overcoming tumor-induced immune suppression. Herein, we review the key properties of nanovaccines for cancer immunotherapy and highlight the recent advances in their development based on the structure and composition of various (including synthetic and semi (biogenic) nanocarriers. Moreover, we discuss tumor cell-derived vaccines (including those based on whole-tumor-cell components, EVs, cell membrane-encapsulated NPs, and hybrid membrane-coated NPs), nanovaccine action mechanisms, and the challenges of immunocancer therapy and their translation to clinical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India.
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India
| | - Lin Wang
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Qilong Cao
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
10
|
Ekwemalor K, Asiamah E, Adjei-Fremah S, Eluka-Okoludoh E, Mulakala B, Osei B, Worku M. Diverse pathogen-associated molecular patterns affect transcription of genes in the toll-like receptor signaling pathway in goat blood. Anim Biotechnol 2023; 34:3729-3738. [PMID: 37226739 DOI: 10.1080/10495398.2023.2214189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS), peptidoglycan (PGN), Polyinosinic-polycytidylic acid (poly I:C), and CpG Oligodeoxynucleotides (ODN) are recognized by Toll-like receptors (TLR). This study aimed to investigate the effect of diverse PAMPs on the transcription of TLR signaling pathway genes in goat blood. Whole blood was collected from 3 female BoerXSpanish goats and treated with the following PAMPs: 10 µg/ml LPS, PGN, CpG ODN (2216), CpG ODN (2006), and 12.5 µg/ml Poly I:C. Blood-treated PBS served as a control. The expression of 84 genes in the human TLR signaling pathway RT2 PCR Array (Qiagen) was evaluated using real-time PCR. Treatment with PBS affected the expression of 74 genes, Poly I:C affected the expression of 40 genes, t ODN 2006 affected the expression of 50 genes, ODN 2216 affected the expression of 52 genes, LPS affected the expression of 49 genes, while PGN affected the expression of 49 genes. Our results show that PAMPs modulated and increased the expression of genes in the TLR signaling pathway. These results highlight important insights into how the host responds to different pathogens and may help design adjuvants for therapeutics and vaccines that target different.
Collapse
Affiliation(s)
- Kingsley Ekwemalor
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Emmanuel Asiamah
- Department of Agriculture, University of Arkansas at Pine Bluff, Pine Bluff, AR, USA
| | - Sarah Adjei-Fremah
- Department of Biological Sciences, Winston-Salem State University, Winston-Salem, NC, USA
| | - Eboghoye Eluka-Okoludoh
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Bharath Mulakala
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Bertha Osei
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Mulumebet Worku
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| |
Collapse
|
11
|
Song S, Wang S, Jiang X, Yang F, Gao S, Lin X, Cheng H, van der Veen S. Th1-polarized MtrE-based gonococcal vaccines display prophylactic and therapeutic efficacy. Emerg Microbes Infect 2023; 12:2249124. [PMID: 37584947 PMCID: PMC10467530 DOI: 10.1080/22221751.2023.2249124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/17/2023]
Abstract
ABSTRACTGlobal dissemination of high-level ceftriaxone-resistant Neisseria gonorrhoeae strains associated with the FC428 clone poses a threat to the efficacy ceftriaxone-based therapies. Vaccination is the best strategy to contain multidrug-resistant infections. In this study, we investigated the efficacy of MtrE and its surface Loop2 as vaccine antigens when combined with a Th1-polarizing adjuvant, which is expected to be beneficial for gonococcal vaccine development. Using in vitro dendritic cell maturation and T cell differentiation assays, CpG1826 was identified as the optimal Th1-polarizing adjuvant for MtrE and Loop2 displayed as linear epitope (Nloop2) or structural epitope (Intraloop2) on a carrier protein. Loop2-based antigens raised strongly Th1-polarized and bactericidal antibody responses in vaccinated mice. Furthermore, the vaccine formulations provided protection against a gonococcal challenge in mouse vaginal tract infection model when provided as prophylactic vaccines. Also, the vaccine formulations accelerated gonococcal clearance when provided as a single therapeutic dose to treat an already established infection, including against a strain associated with the FC428 clone. Therefore, this study demonstrated that MtrE and Loop 2 are effective gonococcal vaccine antigens when combined with the Th1-polarizing CpG1826 adjuvant.
Collapse
Affiliation(s)
- Shuaijie Song
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Shuyi Wang
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaoyun Jiang
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fan Yang
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Shuai Gao
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xu’ai Lin
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hao Cheng
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Stijn van der Veen
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, People’s Republic of China
| |
Collapse
|
12
|
Kalanaky S, Fakharzadeh S, Karimi P, Hafizi M, Jamaati H, Hassanzadeh SM, Khorasani A, Mahdavi M, Nazaran MH. Nanoadjuvants Produced by Advanced Nanochelating Technology in the Inactivated-Severe Acute Respiratory Syndrome Coronavirus-2 Vaccine Formulation: Preliminary Results on Cytokines and IgG Responses. Viral Immunol 2023; 36:409-423. [PMID: 37506342 DOI: 10.1089/vim.2023.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023] Open
Abstract
Despite the great success of vaccines in various infectious diseases, most current vaccines are not effective enough, and on the contrary, clinically approved alum adjuvants cannot induce sufficient immune responses, including a potent cellular immune response to confer protection. In this study, we used Nanochelating Technology to develop novel nanoadjuvants to boost the potency of the alum-adjuvanted inactivated severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccine. BALB/c mice were immunized twice over 2 weeks with different doses of adjuvanted-vaccine formulations and immune responses were assessed. The analysis results of IFN-γ and IL-17 cytokines demonstrated the effectiveness of the nanoadjuvants produced by the Nanochelating Technology in shifting the alum-based vaccine toward a stronger Th1 pattern. In addition, these nanoadjuvants improved IL-2 cytokine response, which shows the efficacy of these novel formulations in inducing specific T lymphocyte proliferation. Using these nanoadjuvants increased IL-10 cytokine secretion that may be representative of a better immunoregulatory impact and may also potentially prevent immunopathology responses. Moreover, specific IgG titer analysis revealed the potency of these nanoadjuvants in improving humoral immune responses. The enzyme-linked immunosorbent assay of receptor-binding domain (RBD)-specific IgG response showed that the developed novel formulations induced strong IgG responses against this protein. This study shows that the nanostructures produced by the Advanced Nanochelating Technology have potent adjuvant effects on alum-based SARS-CoV-2 vaccines to not only compensate for alum weakness in inducing the cellular immune responses by smart regulation of the immune system but also significantly improve the humoral and cellular immune responses simultaneously.
Collapse
Affiliation(s)
- Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Saideh Fakharzadeh
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Pegah Karimi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mehdi Hassanzadeh
- Department of BCG Vaccine Production, Production and Research Complex, Pasteur Institute of Iran, Karaj, Iran
| | - Akbar Khorasani
- Department of FMD Vaccine Production, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Medical Division, Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Mohammad Hassan Nazaran
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
- Owner of Nanochelating Technology and Executive Manager and Chairman of Management Board of Sodour Ahrar Shargh Company, Tehran, Iran
| |
Collapse
|
13
|
Bian Y, Walter DL, Zhang C. Efficiency of Interferon-γ in Activating Dendritic Cells and Its Potential Synergy with Toll-like Receptor Agonists. Viruses 2023; 15:v15051198. [PMID: 37243284 DOI: 10.3390/v15051198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/14/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Interferon-γ (IFN-γ) is a cytokine that plays an important role in immune regulation, especially in the activation and differentiation of immune cells. Toll-like receptors (TLRs) are a family of pattern-recognition receptors that sense structural motifs related to pathogens and alert immune cells to the invasion. Both IFN-γ and TLR agonists have been used as immunoadjuvants to augment the efficacy of cancer immunotherapies and vaccines against infectious diseases or psychoactive compounds. In this study, we aimed to explore the potential of IFN-γ and TLR agonists being applied simultaneously to boost dendritic cell activation and the subsequent antigen presentation. In brief, murine dendritic cells were treated with IFN-γ and/or the TLR agonists, polyinosinic-polycytidylic acid (poly I:C), or resiquimod (R848). Next, the dendritic cells were stained for an activation marker, a cluster of differentiation 86 (CD86), and the percentage of CD86-positive cells was measured by flow cytometry. From the cytometric analysis, IFN-γ efficiently stimulated a considerable number of the dendritic cells, while the TLR agonists by themselves could merely activate a few compared to the control. The combination of IFN-γ with poly I:C or R848 triggered a higher amount of dendritic cell activation than IFN-γ alone. For instance, 10 ng/mL IFN-γ with 100 µg/mL poly I:C achieved 59.1% cell activation, which was significantly higher than the 33.4% CD86-positive cells obtained by 10 ng/mL IFN-γ. These results suggested that IFN-γ and TLR agonists could be applied as complementary systems to promote dendritic cell activation and antigen presentation. There might be a synergy between the two classes of molecules, but further investigation is warranted to ascertain the interaction of their promotive activities.
Collapse
Affiliation(s)
- Yuanzhi Bian
- Department of Biological Systems Engineering, College of Agriculture and Life Sciences & College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Debra L Walter
- Department of Biological Systems Engineering, College of Agriculture and Life Sciences & College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Chenming Zhang
- Department of Biological Systems Engineering, College of Agriculture and Life Sciences & College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
14
|
Butkovich N, Tucker JA, Ramirez A, Li E, Meli VS, Nelson EL, Wang SW. Nanoparticle vaccines can be designed to induce pDC support of mDCs for increased antigen display. Biomater Sci 2023; 11:596-610. [PMID: 36476811 PMCID: PMC10775882 DOI: 10.1039/d2bm01132h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cancer vaccine immunotherapy facilitates the immune system's recognition of tumor-associated antigens, and the biomolecular design of these vaccines using nanoparticles is one important approach towards obtaining strong anti-tumor responses. Following activation of dendritic cells (DCs), a robust CD8+ T cell-mediated adaptive immune response is critical for tumor elimination. While the role of efficient antigen-presenting myeloid DCs (mDCs) is conventionally attributed towards vaccine efficacy, participation by highly cytokine-producing plasmacytoid DCs (pDCs) is less understood and is often overlooked. We examined vaccines based on the E2 protein nanoparticle platform that delivered encapsulated TLR9 agonist bacterial-like DNA (CpG1826 or CpG1018) or TLR7 agonist viral ssRNA to determine their efficacy over free agonists in activating both mDCs and pDCs for antigen presentation. Although mDCs were only activated by nanoparticle-encapsulated TLR9 agonists, pDCs were activated by all the individually tested constructs, and CpG1826 was shown to induce pDC cytokine production. Transfer of secreted factors from pDCs that were stimulated with a vaccine formulation comprising peptide antigen and CpG1826 enhanced mDC display of the antigen, particularly when delivered in nanoparticles. Only when treated with nanoparticle-conjugated vaccine could pDCs secrete factors to induce antigen display on naïve mDCs. These results reveal that pDCs can aid mDCs, highlighting the importance of activating both pDCs and mDCs in designing effective cancer vaccines, and demonstrate the advantage of using nanoparticle-based vaccine delivery.
Collapse
Affiliation(s)
- Nina Butkovich
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Jo Anne Tucker
- Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Aaron Ramirez
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Enya Li
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Edward L Nelson
- Department of Medicine, University of California, Irvine, CA 92697, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
- Institute for Immunology, University of California, Irvine, CA 92697, USA
| | - Szu-Wen Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
- Institute for Immunology, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| |
Collapse
|
15
|
Cedrone E, Dobrovolskaia MA. Detection of Nanoparticles' Ability to Stimulate Toll-Like Receptors Using HEK-Blue Reporter Cell Lines. Methods Mol Biol 2023; 2709:241-251. [PMID: 37572285 DOI: 10.1007/978-1-0716-3417-2_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/14/2023]
Abstract
Nanoparticles can be used to formulate Toll-like receptor (TLR) agonists as vaccine and immunotherapy adjuvants or contain undesirable contaminants (e.g., endotoxin, CpG DNA, flagellin) with TLR-agonist activity. In both scenarios, the activation of the innate immune pattern recognition receptor leads to the inflammatory response that can be beneficial as in the case with vaccines and immunotherapies or adverse as in the case with contaminants. The protocol described herein utilizes commercially available reporter cell lines expressing individual TLRs, which, upon activation with their cognate agonists, stimulate the cells to produce secreted alkaline phosphatase detectable using a plate reader.
Collapse
Affiliation(s)
- Edward Cedrone
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
16
|
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]
|
17
|
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: 24] [Impact Index Per Article: 8.0] [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
|
18
|
Therapeutic applications of toll-like receptors (TLRs) agonists in AML. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 24:2319-2329. [PMID: 35962918 DOI: 10.1007/s12094-022-02917-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive type of blood cancer affecting bone marrow (BM). In AML, hematopoietic precursors are arrested in the early stages of development and are defined as the presence of ≥ 20% blasts (leukemia cells) in the BM. Toll-like receptors (TLR) are major groups of pattern recognition receptors expressed by almost all innate immune cells that enable them to detect a wide range of pathogen-associated molecular patterns and damage-associated molecular patterns to prime immune responses toward adaptive immunity. Because TLRs are commonly expressed on transformed immune system cells (ranging from blasts to memory cells), they can be a potential option for developing efficient clinical alternatives in hematologic tumors. This is because several in vitro and in vivo investigations have demonstrated that TLR signaling increased the immunogenicity of AML cells, making them more vulnerable to T cell-mediated invasion. This study aimed to review the current knowledge in this field and provide some insight into the therapeutic potentials of TLRs in AML.
Collapse
|
19
|
Fan J, Jin S, Gilmartin L, Toth I, Hussein WM, Stephenson RJ. Advances in Infectious Disease Vaccine Adjuvants. Vaccines (Basel) 2022; 10:1120. [PMID: 35891284 PMCID: PMC9316175 DOI: 10.3390/vaccines10071120] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Vaccines are one of the most significant medical interventions in the fight against infectious diseases. Since their discovery by Edward Jenner in 1796, vaccines have reduced the worldwide transmission to eradication levels of infectious diseases, including smallpox, diphtheria, hepatitis, malaria, and influenza. However, the complexity of developing safe and effective vaccines remains a barrier for combating many more infectious diseases. Immune stimulants (or adjuvants) are an indispensable factor in vaccine development, especially for inactivated and subunit-based vaccines due to their decreased immunogenicity compared to whole pathogen vaccines. Adjuvants are widely diverse in structure; however, their overall function in vaccine constructs is the same: to enhance and/or prolong an immunological response. The potential for adverse effects as a result of adjuvant use, though, must be acknowledged and carefully managed. Understanding the specific mechanisms of adjuvant efficacy and safety is a key prerequisite for adjuvant use in vaccination. Therefore, rigorous pre-clinical and clinical research into adjuvant development is essential. Overall, the incorporation of adjuvants allows for greater opportunities in advancing vaccine development and the importance of immune stimulants drives the emergence of novel and more effective adjuvants. This article highlights recent advances in vaccine adjuvant development and provides detailed data from pre-clinical and clinical studies specific to infectious diseases. Future perspectives into vaccine adjuvant development are also highlighted.
Collapse
Affiliation(s)
- Jingyi Fan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Shengbin Jin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Lachlan Gilmartin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Rachel J. Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| |
Collapse
|
20
|
Kaur A, Baldwin J, Brar D, Salunke DB, Petrovsky N. Toll-like receptor (TLR) agonists as a driving force behind next-generation vaccine adjuvants and cancer therapeutics. Curr Opin Chem Biol 2022; 70:102172. [PMID: 35785601 DOI: 10.1016/j.cbpa.2022.102172] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 05/18/2022] [Indexed: 01/06/2023]
Abstract
Until recently, the development of new human adjuvants was held back by a poor understanding of their mechanisms of action. The field was revolutionized by the discovery of the toll-like receptors (TLRs), innate immune receptors that directly or indirectly are responsible for detecting pathogen-associated molecular patterns (PAMPs) and respond to them by activating innate and adaptive immune pathways. Hundreds of ligands targeting various TLRs have since been identified and characterized as vaccine adjuvants. This work has important implications not only for the development of vaccines against infectious diseases but also for immuno-therapies against cancer, allergy, Alzheimer's disease, drug addiction and other diseases. Each TLR has its own specific tissue localization and downstream gene signalling pathways, providing researchers the opportunity to precisely tailor adjuvants with specific immune effects. TLR agonists can be combined with other TLR or alternative adjuvants to create combination adjuvants with synergistic or modulatory effects. This review provides an introduction to the various classes of TLR adjuvants and their respective signalling pathways. It provides an overview of recent advancements in the TLR field in the past 2-3 years and discusses criteria for selecting specific TLR adjuvants based on considerations, such as disease mechanisms and correlates of protection, TLR immune biasing capabilities, route of administration, antigen compatibility, new vaccine technology platforms, and age- and species-specific effects.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | | | - Deshkanwar Brar
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, Adelaide 5042, Australia; College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia.
| |
Collapse
|
21
|
Juneja M, Suthar T, Pardhi VP, Ahmad J, Jain K. Emerging trends and promises of nanoemulsions in therapeutics of infectious diseases. Nanomedicine (Lond) 2022; 17:793-812. [PMID: 35587031 DOI: 10.2217/nnm-2022-0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Infectious diseases are prevalent and have contributed to high morbidity rates by creating havoc like the COVID-19, 1918 influenza and Black Death (the plague) pandemics. Antimicrobial resistance, adverse effects, the emergence of co-infections and the high cost of antimicrobial therapies are major threats to the health of people worldwide while impacting overall healthcare and socioeconomic development. One of the most common ways to address this issue lies in improving existing antimicrobial drug-delivery systems. Nanoemulsions and their modified forms have been successfully employed for the delivery of antimicrobials to treat infectious diseases. In this article, the authors comprehensively reviewed how nanoemulsion-based formulation systems are shifting the paradigm for therapeutics and diagnosis of infectious diseases.
Collapse
Affiliation(s)
- Mehak Juneja
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Uttar Pradesh, 226002, India
| | - Teeja Suthar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Uttar Pradesh, 226002, India
| | - Vishwas P Pardhi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Uttar Pradesh, 226002, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, 11001, Saudi Arabia
| | - Keerti Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Uttar Pradesh, 226002, India
| |
Collapse
|
22
|
Stickdorn J, Stein L, Arnold-Schild D, Hahlbrock J, Medina-Montano C, Bartneck J, Ziß T, Montermann E, Kappel C, Hobernik D, Haist M, Yurugi H, Raabe M, Best A, Rajalingam K, Radsak MP, David SA, Koynov K, Bros M, Grabbe S, Schild H, Nuhn L. Systemically Administered TLR7/8 Agonist and Antigen-Conjugated Nanogels Govern Immune Responses against Tumors. ACS NANO 2022; 16:4426-4443. [PMID: 35103463 PMCID: PMC8945363 DOI: 10.1021/acsnano.1c10709] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The generation of specific humoral and cellular immune responses plays a pivotal role in the development of effective vaccines against tumors. Especially the presence of antigen-specific, cytotoxic T cells influences the outcome of therapeutic cancer vaccinations. Different strategies, ranging from delivering antigen-encoding mRNAs to peptides or full antigens, are accessible but often suffer from insufficient immunogenicity and require immune-boosting adjuvants as well as carrier platforms to ensure stability and adequate retention. Here, we introduce a pH-responsive nanogel platform as a two-component antitumor vaccine that is safe for intravenous application and elicits robust immune responses in vitro and in vivo. The underlying chemical design allows for straightforward covalent attachment of a model antigen (ovalbumin) and an immune adjuvant (imidazoquinoline-type TLR7/8 agonist) onto the same nanocarrier system. In addition to eliciting antigen-specific T and B cell responses that outperform mixtures of individual components, our two-component nanovaccine leads in prophylactic and therapeutic studies to an antigen-specific growth reduction of different tumors expressing ovalbumin intracellularly or on their surface. Regarding the versatile opportunities for functionalization, our nanogels are promising for the development of highly customized and potent nanovaccines.
Collapse
Affiliation(s)
- Judith Stickdorn
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Lara Stein
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Danielle Arnold-Schild
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Jennifer Hahlbrock
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Carolina Medina-Montano
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Joschka Bartneck
- III Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University
Mainz, Langenbeckstraße
1, 55131 Mainz, Germany
| | - Tanja Ziß
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Evelyn Montermann
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Cinja Kappel
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Dominika Hobernik
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Maximilian Haist
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Hajime Yurugi
- Cell
Biology Unit, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Marco Raabe
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Andreas Best
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Krishnaraj Rajalingam
- Cell
Biology Unit, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Markus P. Radsak
- III Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University
Mainz, Langenbeckstraße
1, 55131 Mainz, Germany
| | - Sunil A. David
- ViroVax,
LLC, 2029 Becker Drive
Suite 100E, Lawrence 66047-1620, Kansas. United States
| | - Kaloian Koynov
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Matthias Bros
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stephan Grabbe
- Department
of Dermatology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Hansjörg Schild
- Institute
of Immunology, University Medical Center
of Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Lutz Nuhn
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
23
|
Antonelli ACB, Almeida VP, de Castro FOF, Silva JM, Pfrimer IAH, Cunha-Neto E, Maranhão AQ, Brígido MM, Resende RO, Bocca AL, Fonseca SG. In silico construction of a multiepitope Zika virus vaccine using immunoinformatics tools. Sci Rep 2022; 12:53. [PMID: 34997041 PMCID: PMC8741764 DOI: 10.1038/s41598-021-03990-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 12/01/2021] [Indexed: 01/02/2023] Open
Abstract
Zika virus (ZIKV) is an arbovirus from the Flaviviridae family and Flavivirus genus. Neurological events have been associated with ZIKV-infected individuals, such as Guillain-Barré syndrome, an autoimmune acute neuropathy that causes nerve demyelination and can induce paralysis. With the increase of ZIKV infection incidence in 2015, malformation and microcephaly cases in newborns have grown considerably, which suggested congenital transmission. Therefore, the development of an effective vaccine against ZIKV became an urgent need. Live attenuated vaccines present some theoretical risks for administration in pregnant women. Thus, we developed an in silico multiepitope vaccine against ZIKV. All structural and non-structural proteins were investigated using immunoinformatics tools designed for the prediction of CD4 + and CD8 + T cell epitopes. We selected 13 CD8 + and 12 CD4 + T cell epitopes considering parameters such as binding affinity to HLA class I and II molecules, promiscuity based on the number of different HLA alleles that bind to the epitopes, and immunogenicity. ZIKV Envelope protein domain III (EDIII) was added to the vaccine construct, creating a hybrid protein domain-multiepitope vaccine. Three high scoring continuous and two discontinuous B cell epitopes were found in EDIII. Aiming to increase the candidate vaccine antigenicity even further, we tested secondary and tertiary structures and physicochemical parameters of the vaccine conjugated to four different protein adjuvants: flagellin, 50S ribosomal protein L7/L12, heparin-binding hemagglutinin, or RS09 synthetic peptide. The addition of the flagellin adjuvant increased the vaccine's predicted antigenicity. In silico predictions revealed that the protein is a probable antigen, non-allergenic and predicted to be stable. The vaccine’s average population coverage is estimated to be 87.86%, which indicates it can be administered worldwide. Peripheral Blood Mononuclear Cells (PBMC) of individuals with previous ZIKV infection were tested for cytokine production in response to the pool of CD4 and CD8 ZIKV peptide selected. CD4 + and CD8 + T cells showed significant production of IFN-γ upon stimulation and IL-2 production was also detected by CD8 + T cells, which indicated the potential of our peptides to be recognized by specific T cells and induce immune response. In conclusion, we developed an in silico universal vaccine predicted to induce broad and high-coverage cellular and humoral immune responses against ZIKV, which can be a good candidate for posterior in vivo validation.
Collapse
Affiliation(s)
- Ana Clara Barbosa Antonelli
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Rua 235 s/n, sala 335, Setor Universitário, Goiânia, GO, 74605-050, Brazil
| | - Vinnycius Pereira Almeida
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Rua 235 s/n, sala 335, Setor Universitário, Goiânia, GO, 74605-050, Brazil
| | - Fernanda Oliveira Feitosa de Castro
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Rua 235 s/n, sala 335, Setor Universitário, Goiânia, GO, 74605-050, Brazil.,Departament of Master in Environmental Sciences and Health, School of Medical, Pharmaceutical and Biomedical Sciences, Pontifical Catholic University of Goiás, Goiânia, Brazil
| | | | - Irmtraut Araci Hoffmann Pfrimer
- Departament of Master in Environmental Sciences and Health, School of Medical, Pharmaceutical and Biomedical Sciences, Pontifical Catholic University of Goiás, Goiânia, Brazil
| | - Edecio Cunha-Neto
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii) - National Institute of Science and Technology (INCT), São Paulo, Brazil
| | - Andréa Queiroz Maranhão
- Department of Cell Biology, University of Brasília, Brasília, Brazil.,Institute for Investigation in Immunology (iii) - National Institute of Science and Technology (INCT), São Paulo, Brazil
| | - Marcelo Macedo Brígido
- Department of Cell Biology, University of Brasília, Brasília, Brazil.,Institute for Investigation in Immunology (iii) - National Institute of Science and Technology (INCT), São Paulo, Brazil
| | | | | | - Simone Gonçalves Fonseca
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Rua 235 s/n, sala 335, Setor Universitário, Goiânia, GO, 74605-050, Brazil. .,Institute for Investigation in Immunology (iii) - National Institute of Science and Technology (INCT), São Paulo, Brazil.
| |
Collapse
|
24
|
Božić Nedeljković B, Ćilerdžić J, Zmijanjac D, Marković M, Džopalić T, Vasilijić S, Stajić M, Vučević D. Immunomodulatory effects of extract of Lingzhi or Reishi medicinal Mushroom Ganoderma lucidum (Agaricomycetes) basidiocarps cultivated on alternative substrate. Int J Med Mushrooms 2022; 24:45-59. [DOI: 10.1615/intjmedmushrooms.2022044452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
25
|
Swartz AM, Hotchkiss KM, Nair SK, Sampson JH, Batich KA. Generation of Tumor Targeted Dendritic Cell Vaccines with Improved Immunogenic and Migratory Phenotype. Methods Mol Biol 2022; 2410:609-626. [PMID: 34914072 DOI: 10.1007/978-1-0716-1884-4_33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Our group has employed methodologies for effective ex vivo generation of dendritic cell (DC) vaccines for patients with primary malignant brain tumors. In order to reliably produce the most potent, most representational vaccinated DC that will engender an antitumor response requires the ability to orchestrate multiple methodologies that address antigen cross-presentation, T-cell costimulation and polarization, and migratory capacity. In this chapter, we describe a novel method for augmenting the immunogenicity and migratory potential of DCs for their use as vaccines. We have elucidated methodologies to avoid the phenomenon known as immunodominance in generating cancer vaccines. We have found that culturing DC progenitors in serum-free conditions for the duration of the differentiation protocol results in a more homogeneously mature population of DCs that exhibit enhanced immunogenicity compared to DCs generated in serum-containing culture conditions. Furthermore, we demonstrate our method for generating high mobility DCs that readily migrate toward lymphoid organ chemoattractants using CCL3 protein. The combination of these two approaches represents a facile and clinically tractable methodology for generating highly mature DCs with excellent migratory capacity.
Collapse
Affiliation(s)
- Adam M Swartz
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Kelly M Hotchkiss
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Smita K Nair
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC, USA
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| | - John H Sampson
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| | - Kristen A Batich
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
26
|
Zhou Q, Zhou R, Yang H, Yang H. To Be or Not To Be Vaccinated: That Is a Question in Myasthenia Gravis. Front Immunol 2021; 12:733418. [PMID: 34603311 PMCID: PMC8485039 DOI: 10.3389/fimmu.2021.733418] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/01/2021] [Indexed: 01/04/2023] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease characterized by muscle weakness and abnormal fatigability due to the antibodies against postsynaptic receptors. Despite the individual discrepancy, patients with MG share common muscle weakness, autoimmune dysfunction, and immunosuppressive treatment, which predispose them to infections that can trigger or exacerbate MG. Vaccination, as a mainstay of prophylaxis, is a major management strategy. However, the past years have seen growth in vaccine hesitancy, owing to safety and efficacy concerns. Ironically, vaccines, serving as an essential and effective means of defense, may induce similar immune cross-reactivity to what they are meant to prevent. Herein, we outline the progress in vaccination, review the current status, and postulate the clinical association among MG, vaccination, and immunosuppression. We also address safety and efficacy concerns of vaccination in MG, in relation to COVID-19. Since only a handful of studies have reported vaccination in individuals with MG, we further review the current clinical studies and guidelines in rheumatic diseases. Overall, our reviews offer a reference to guide future vaccine clinical decision-making and improve the management of MG patients.
Collapse
Affiliation(s)
| | | | | | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
Yamaguchi T, Yoshimura T, Ohara T, Fujisawa M, Tong G, Matsukawa A. PolyI:C suppresses TGF-β1-induced Akt phosphorylation and reduces the motility of A549 lung carcinoma cells. Mol Biol Rep 2021; 48:6313-6321. [PMID: 34390443 DOI: 10.1007/s11033-021-06625-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/04/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUNDS Epithelial mesenchymal transition (EMT) is a critical process involved in the invasion and metastasis of cancer, including lung cancer (LC). Transforming growth factor (TGF)-β is one of factors capable of inducing EMT. Polyinosinic-polycytidylic acid (polyI:C), a synthetic agonist for toll-like receptor (TLR) 3, can enhance immune responses and has been used as an adjuvant for cancer vaccines; however, it remains unclear whether it influences other process, such as EMT. In the present study, we examined the effects of polyI:C on TGF-β-treated A549 human LC cells. METHODS AND RESULTS By in vitro cell proliferation assay, polyI:C showed no effect on the growth of A549 cells treated with TGF-β1 at the concentration range up to 10 μg/ml; however, it markedly suppressed the motility in a cell scratch and a cell invasion assay. By Western blotting, polyI:C dramatically decreased TGF-β1-induced Ak strain transforming (Akt) phosphorylation and increased phosphatase and tensin homologue (PTEN) expression without affecting the Son of mothers against decapentaplegic (Smad) 3 phosphorylation or the expression level of E-cadherin, N-cadherin or Snail, indicating that polyI:C suppressed cell motility independently of the 'cadherin switching'. The Akt inhibitor perifosine inhibited TGF-β1-induced cell invasion, and the PTEN-specific inhibitor VO-OHpic appeared to reverse the inhibitory effect of polyI:C. CONCLUSION PolyI:C has a novel function to suppress the motility of LC cells undergoing EMT by targeting the phosphatidylinositol 3-kinase/Akt pathway partly via PTEN and may prevent or reduce the metastasis of LC cells.
Collapse
Affiliation(s)
- Takahiro Yamaguchi
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Masayoshi Fujisawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Gao Tong
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
28
|
Wijesinghe VN, Farouk IA, Zabidi NZ, Puniyamurti A, Choo WS, Lal SK. Current vaccine approaches and emerging strategies against herpes simplex virus (HSV). Expert Rev Vaccines 2021; 20:1077-1096. [PMID: 34296960 DOI: 10.1080/14760584.2021.1960162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Vaccine development for the disease caused by the herpes simplex virus (HSV) has been challenging over the years and is always in dire need of novel approaches for prevention and cure. To date, the HSV disease remains incurable and challenging to prevent. The disease is extremely widespread due to its high infection rate, resulting in millions of infection cases worldwide.Areas covered: This review first explains the diverse forms of HSV-related disease presentations and reports past vaccine history for the disease. Next, this review examines current and novel HSV vaccine approaches being studied and tested for efficacy and safety as well as vaccines in clinical trial phases I to III. Modern approaches to vaccine design using bioinformatics are described. Finally, we discuss measures to enhance new vaccine development pipelines for HSV.Expert opinion: Modernized approaches using in silico analysis and bioinformatics are emerging methods that exhibit potential for producing vaccines with enhanced targets and formulations. Although not yet fully established for HSV disease, we describe current studies using these approaches for HSV vaccine design to shed light on these methods. In addition, we provide up-to-date requirements of immunogenicity, adjuvant selection, and routes of administration.
Collapse
Affiliation(s)
| | - Isra Ahmad Farouk
- School of Science, Monash University, Bandar Sunway, Selangor, Malaysia
| | | | | | - Wee Sim Choo
- School of Science, Monash University, Bandar Sunway, Selangor, Malaysia
| | - Sunil Kumar Lal
- School of Science, Monash University, Bandar Sunway, Selangor, Malaysia.,Tropical Medicine & Biology Platform, Monash University, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
29
|
Najem H, Khasraw M, Heimberger AB. Immune Microenvironment Landscape in CNS Tumors and Role in Responses to Immunotherapy. Cells 2021; 10:2032. [PMID: 34440802 PMCID: PMC8393758 DOI: 10.3390/cells10082032] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the important evolution of immunotherapeutic agents, brain tumors remain, in general, refractory to immune therapeutics. Recent discoveries have revealed that the glioma microenvironment includes a wide variety of immune cells in various states that play an important role in the process of tumorigenesis. Anti-tumor immune activity may be occurring or induced in immunogenic hot spots or at the invasive edge of central nervous system (CNS) tumors. Understanding the complex heterogeneity of the immune microenvironment in gliomas will likely be the key to unlocking the full potential of immunotherapeutic strategies. An essential consideration will be the induction of immunological effector responses in the setting of the numerous aspects of immunosuppression and evasion. As such, immune therapeutic combinations are a fundamental objective for clinical studies in gliomas. Through immune profiling conducted on immune competent murine models of glioma and ex vivo human glioma tissue, we will discuss how the frequency, distribution of immune cells within the microenvironment, and immune modulatory processes, may be therapeutically modulated to lead to clinical benefits.
Collapse
Affiliation(s)
- Hinda Najem
- Department of Neurological Surgery and Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA;
| | - Amy B. Heimberger
- Department of Neurological Surgery and Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| |
Collapse
|
30
|
Melssen MM, Pollack KE, Meneveau MO, Smolkin ME, Pinczewski J, Koeppel AF, Turner SD, Sol-Church K, Hickman A, Deacon DH, Petroni GR, Slingluff CL. Characterization and comparison of innate and adaptive immune responses at vaccine sites in melanoma vaccine clinical trials. Cancer Immunol Immunother 2021; 70:2151-2164. [PMID: 33454795 PMCID: PMC10992166 DOI: 10.1007/s00262-020-02844-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022]
Abstract
The strength and durability of systemic anti-tumor immune responses induced by cancer vaccines depends on adjuvants to support an immunogenic vaccine site microenvironment (VSME). Adjuvants include water-in-oil emulsions with incomplete Freund's adjuvant (IFA) and combinations of toll-like receptor (TLR) agonists, including a preparation containing TLR4 and TLR9 agonists with QS-21 (AS15). IFA-containing vaccines can promote immune cell accumulation at the VSME, whereas effects of AS15 are largely unexplored. Therefore, we assessed innate and adaptive immune cell accumulation and gene expression at the VSME after vaccination with AS15 and compared to effects with IFA. We hypothesized that AS15 would promote less accumulation of innate and adaptive immune cells at the VSME than IFA vaccines. In two clinical trials, patients with resected high-risk melanoma received either a multipeptide vaccine with IFA or a recombinant MAGE-A3 protein vaccine with AS15. Vaccine site biopsies were obtained after one or multiple vaccines. T cells accumulated early after vaccines with AS15, but this was not durable or of the same magnitude as vaccination in IFA. Vaccines with AS15 increased durable expression of DC- and T cell-related genes, as well as PD-L1 and IDO1, suggesting complex activation and regulation of innate and adaptive immune function with AS15. These changes were generally greater with vaccines containing IFA, but IFA induced reduction in myeloid suppressor cells markers. Evidence of tertiary lymphoid structure (TLS) formation was observed with both adjuvants. Our findings highlight adjuvant-dependent changes in immune features at the VSME that may impact systemic immune responses.
Collapse
Affiliation(s)
- Marit M Melssen
- Department of Surgery, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Karlyn E Pollack
- Department of Surgery, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Max O Meneveau
- Department of Surgery, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Mark E Smolkin
- Department of Public Health Sciences, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Joel Pinczewski
- Department of Surgery, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Alexander F Koeppel
- Department of Public Health Sciences, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Stephen D Turner
- Department of Public Health Sciences, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Katia Sol-Church
- Department of Pathology, University of Virginia, Charlottesville, USA
| | - Alexandra Hickman
- Department of Surgery, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Donna H Deacon
- Department of Surgery, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Gina R Petroni
- Department of Public Health Sciences, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA
| | - Craig L Slingluff
- Department of Surgery, University of Virginia, P.O. Box 801329, Charlottesville, VA, 22908, USA.
| |
Collapse
|
31
|
Abstract
A growing theme in chemistry is the joining of multiple organic molecular building blocks to create functional molecules. Diverse derivatizable structures—here termed “scaffolds” comprised of “hubs”—provide the foundation for systematic covalent organization of a rich variety of building blocks. This review encompasses 30 tri- or tetra-armed molecular hubs (e.g., triazine, lysine, arenes, dyes) that are used directly or in combination to give linear, cyclic, or branched scaffolds. Each scaffold is categorized by graph theory into one of 31 trees to express the molecular connectivity and overall architecture. Rational chemistry with exacting numbers of derivatizable sites is emphasized. The incorporation of water-solubilization motifs, robust or self-immolative linkers, enzymatically cleavable groups and functional appendages affords immense (and often late-stage) diversification of the scaffolds. Altogether, 107 target molecules are reviewed along with 19 syntheses to illustrate the distinctive chemistries for creating and derivatizing scaffolds. The review covers the history of the field up through 2020, briefly touching on statistically derivatized carriers employed in immunology as counterpoints to the rationally assembled and derivatized scaffolds here, although most citations are from the past two decades. The scaffolds are used widely in fields ranging from pure chemistry to artificial photosynthesis and biomedical sciences.
Collapse
|
32
|
Kash N, Silapunt S. A review of emerging and non-US FDA-approved topical agents for the treatment of basal cell carcinoma. Future Oncol 2021; 17:3111-3132. [PMID: 34156307 DOI: 10.2217/fon-2020-1147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although surgical therapy continues to be the gold standard for the treatment of basal cell carcinoma given high cure rates and the ability to histologically confirm tumor clearance, there are a number of nonsurgical treatment options that may be considered based on individual tumor characteristics, functional and cosmetic considerations, patient comorbidities and patient preference. Topical 5-fluorouracil 5% cream and imiquimod 5% cream have been US FDA-approved for the treatment of superficial basal cell carcinoma. Additionally, a number of new and emerging topical agents and techniques have been described for the treatment of basal cell carcinoma and will be reviewed herein.
Collapse
Affiliation(s)
- Natalie Kash
- Department of Dermatology, Kansas City University-Graduate Medical Education Consortium/Advanced Dermatology & Cosmetic Surgery Orlando Program, Maitland, FL 32751, USA
| | - Sirunya Silapunt
- Department of Dermatology, University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
| |
Collapse
|
33
|
Jin SM, Lee SN, Yoo YJ, Lim YT. Molecular and Macroscopic Therapeutic Systems for Cytokine‐Based Cancer Immunotherapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and Biomedical Institute for Convergence at SKKU Sungkyunkwan University 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and Biomedical Institute for Convergence at SKKU Sungkyunkwan University 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Yeon Jeong Yoo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and Biomedical Institute for Convergence at SKKU Sungkyunkwan University 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and Biomedical Institute for Convergence at SKKU Sungkyunkwan University 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Chemical Engineering Sungkyunkwan University 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| |
Collapse
|
34
|
Chuntova P, Chow F, Watchmaker PB, Galvez M, Heimberger AB, Newell EW, Diaz A, DePinho RA, Li MO, Wherry EJ, Mitchell D, Terabe M, Wainwright DA, Berzofsky JA, Herold-Mende C, Heath JR, Lim M, Margolin KA, Chiocca EA, Kasahara N, Ellingson BM, Brown CE, Chen Y, Fecci PE, Reardon DA, Dunn GP, Liau LM, Costello JF, Wick W, Cloughesy T, Timmer WC, Wen PY, Prins RM, Platten M, Okada H. Unique challenges for glioblastoma immunotherapy-discussions across neuro-oncology and non-neuro-oncology experts in cancer immunology. Meeting Report from the 2019 SNO Immuno-Oncology Think Tank. Neuro Oncol 2021; 23:356-375. [PMID: 33367885 PMCID: PMC7992879 DOI: 10.1093/neuonc/noaa277] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy has made remarkable advances with over 50 separate Food and Drug Administration (FDA) approvals as first- or second-line indications since 2015. These include immune checkpoint blocking antibodies, chimeric antigen receptor-transduced T cells, and bispecific T-cell-engaging antibodies. While multiple cancer types now benefit from these immunotherapies, notable exceptions thus far include brain tumors, such as glioblastoma. As such, it seems critical to gain a better understanding of unique mechanistic challenges underlying the resistance of malignant gliomas to immunotherapy, as well as to acquire insights into the development of future strategies. An Immuno-Oncology Think Tank Meeting was held during the 2019 Annual Society for Neuro-Oncology Scientific Conference. Discussants in the fields of neuro-oncology, neurosurgery, neuro-imaging, medical oncology, and cancer immunology participated in the meeting. Sessions focused on topics such as the tumor microenvironment, myeloid cells, T-cell dysfunction, cellular engineering, and translational aspects that are critical and unique challenges inherent with primary brain tumors. In this review, we summarize the discussions and the key messages from the meeting, which may potentially serve as a basis for advancing the field of immune neuro-oncology in a collaborative manner.
Collapse
Affiliation(s)
- Pavlina Chuntova
- Department of Neurological Surgery, UCSF, San Francisco, California
| | - Frances Chow
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Mildred Galvez
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, Los Angeles, California
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Aaron Diaz
- Department of Neurological Surgery, UCSF, San Francisco, California
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - E John Wherry
- Department of Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Duane Mitchell
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, Florida
| | - Masaki Terabe
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jay A Berzofsky
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | | | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kim A Margolin
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Benjamin M Ellingson
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Christine E Brown
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Yvonne Chen
- Department of Microbiology, Immunology & Molecular Genetics, UCLA, Los Angeles, California
| | - Peter E Fecci
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina
| | - David A Reardon
- Department of Medicine/Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Timothy Cloughesy
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - William C Timmer
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Robert M Prins
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, Los Angeles, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany.,DKTK CCU Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hideho Okada
- Department of Neurological Surgery, UCSF, San Francisco, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| |
Collapse
|
35
|
Blaauboer A, Sideras K, van Eijck CHJ, Hofland LJ. Type I interferons in pancreatic cancer and development of new therapeutic approaches. Crit Rev Oncol Hematol 2020; 159:103204. [PMID: 33387625 DOI: 10.1016/j.critrevonc.2020.103204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 12/17/2020] [Accepted: 12/20/2020] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has emerged as a new treatment strategy for cancer. However, its promise in pancreatic cancer has not yet been realized. Understanding the immunosuppressive tumor microenvironment of pancreatic cancer, and identifying new therapeutic targets to increase tumor-specific immune responses, is necessary in order to improve clinical outcomes. Type I interferons, e.g. IFN-α and -β, are considered as an important bridge between the innate and adaptive immune system. Thereby, type I IFNs induce a broad spectrum of anti-tumor effects, including immunologic, vascular, as well as direct anti-tumor effects. While IFN therapies have been around for a while, new insights into exogenous and endogenous activation of the IFN pathway have resulted in new IFN-related cancer treatment strategies. Here, we focus on the pre-clinical and clinical evidence of novel ways to take advantage of the type I IFN pathway, such as IFN based conjugates and activation of the STING and RIG-I pathways.
Collapse
Affiliation(s)
- Amber Blaauboer
- Department of Surgery, Rotterdam, The Netherlands; Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Leo J Hofland
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Schmitt S, Tahk S, Lohner A, Hänel G, Maiser A, Hauke M, Patel L, Rothe M, Josenhans C, Leonhardt H, Griffioen M, Deiser K, Fenn NC, Hopfner KP, Subklewe M. Fusion of Bacterial Flagellin to a Dendritic Cell-Targeting αCD40 Antibody Construct Coupled With Viral or Leukemia-Specific Antigens Enhances Dendritic Cell Maturation and Activates Peptide-Responsive T Cells. Front Immunol 2020; 11:602802. [PMID: 33281829 PMCID: PMC7689061 DOI: 10.3389/fimmu.2020.602802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/19/2020] [Indexed: 12/30/2022] Open
Abstract
Conventional dendritic cell (DC) vaccine strategies, in which DCs are loaded with antigens ex vivo, suffer biological issues such as impaired DC migration capacity and laborious GMP production procedures. In a promising alternative, antigens are targeted to DC-associated endocytic receptors in vivo with antibody–antigen conjugates co-administered with toll-like receptor (TLR) agonists as adjuvants. To combine the potential advantages of in vivo targeting of DCs with those of conjugated TLR agonists, we generated a multifunctional antibody construct integrating the DC-specific delivery of viral- or tumor-associated antigens and DC activation by TLR ligation in one molecule. We validated its functionality in vitro and determined if TLR ligation might improve the efficacy of such a molecule. In proof-of-principle studies, an αCD40 antibody containing a CMV pp65-derived peptide as an antigen domain (αCD40CMV) was genetically fused to the TLR5-binding D0/D1 domain of bacterial flagellin (αCD40.FlgCMV). The analysis of surface maturation markers on immature DCs revealed that fusion of flagellin to αCD40CMV highly increased DC maturation (3.4-fold elevation of CD80 expression compared to αCD40CMV alone) by specifically interacting with TLR5. Immature DCs loaded with αCD40.FlgCMV induced significantly higher CMVNLV-specific T cell activation and proliferation compared to αCD40CMV in co-culture experiments with allogeneic and autologous T cells (1.8-fold increase in % IFN-γ/TNF-α+ CD8+ T cells and 3.9-fold increase in % CMVNLV-specific dextramer+ CD8+ T cells). More importantly, we confirmed the beneficial effects of flagellin-dependent DC stimulation using a tumor-specific neoantigen as the antigen domain. Specifically, the acute myeloid leukemia (AML)-specific mutated NPM1 (mNPM1)-derived neoantigen CLAVEEVSL was delivered to DCs in the form of αCD40mNPM1 and αCD40.FlgmNPM1 antibody constructs, making this study the first to investigate mNPM1 in a DC vaccination context. Again, αCD40.FlgmNPM1-loaded DCs more potently activated allogeneic mNPM1CLA-specific T cells compared to αCD40mNPM1. These in vitro results confirmed the functionality of our multifunctional antibody construct and demonstrated that TLR5 ligation improved the efficacy of the molecule. Future mouse studies are required to examine the T cell-activating potential of αCD40.FlgmNPM1 after targeting of dendritic cells in vivo using AML xenograft models.
Collapse
Affiliation(s)
- Saskia Schmitt
- Gene Center and Department of Biochemistry, Ludwig Maximilians University Munich, Munich, Germany
| | - Siret Tahk
- Gene Center and Department of Biochemistry, Ludwig Maximilians University Munich, Munich, Germany
| | - Alina Lohner
- Department of Medicine III, University Hospital, Ludwig Maximilians University Munich, Munich, Germany.,Gene Center Munich, Laboratory for Translational Cancer Immunology, Ludwig Maximilians University Munich, Munich, Germany
| | - Gerulf Hänel
- Department of Medicine III, University Hospital, Ludwig Maximilians University Munich, Munich, Germany.,Gene Center Munich, Laboratory for Translational Cancer Immunology, Ludwig Maximilians University Munich, Munich, Germany
| | - Andreas Maiser
- Department of Biology II, Center for Integrated Protein Science, Ludwig Maximilians University Munich, Munich, Germany
| | - Martina Hauke
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, Munich, Germany
| | - Lubna Patel
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, Munich, Germany
| | - Maurine Rothe
- Department of Medicine III, University Hospital, Ludwig Maximilians University Munich, Munich, Germany.,Gene Center Munich, Laboratory for Translational Cancer Immunology, Ludwig Maximilians University Munich, Munich, Germany
| | - Christine Josenhans
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, Munich, Germany.,German Center of Infection Research, DZIF, Munich, Germany
| | - Heinrich Leonhardt
- Department of Biology II, Center for Integrated Protein Science, Ludwig Maximilians University Munich, Munich, Germany
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Katrin Deiser
- Department of Medicine III, University Hospital, Ludwig Maximilians University Munich, Munich, Germany.,Gene Center Munich, Laboratory for Translational Cancer Immunology, Ludwig Maximilians University Munich, Munich, Germany
| | - Nadja C Fenn
- Gene Center and Department of Biochemistry, Ludwig Maximilians University Munich, Munich, Germany
| | - Karl-Peter Hopfner
- Gene Center and Department of Biochemistry, Ludwig Maximilians University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilians University Munich, Munich, Germany.,Gene Center Munich, Laboratory for Translational Cancer Immunology, Ludwig Maximilians University Munich, Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
37
|
Lee SN, Jin SM, Shin HS, Lim YT. Chemical Strategies to Enhance the Therapeutic Efficacy of Toll-like Receptor Agonist Based Cancer Immunotherapy. Acc Chem Res 2020; 53:2081-2093. [PMID: 32966047 DOI: 10.1021/acs.accounts.0c00337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent developments in the fields of biomedical chemistry and immune bioengineering have enabled innovative therapeutic approaches that can enhance the efficacy, accuracy, and safety of cancer immunotherapy. Among the numerous strategies utilized in cancer immunotherapy, Toll-like receptor (TLR) agonist-based approaches have been studied for a long time since they trigger the innate immune system and generate antigen-specific T cell responses to fight against tumors. In addition to these immunostimulatory functions, TLR agonists also contribute to the reprogramming of immune suppressive tumor microenvironments. Although TLR agonists are now being intensively studied in clinical trials due to their substantial immunomodulatory properties, they still show a low therapeutic index. Nonspecific and random stimulation of various immune cells produces excess levels of proinflammatory cytokines, resulting in cytokine storms and chronic diseases. Therefore, the development of chemical strategies to enhance the therapeutic efficacy as well as the safety of TLR agonist-based immunotherapy is essential and in high demand.In this Account, we summarize and discuss recent developments in biomedical chemistry and bioengineering techniques for the immunomodulation of TLR agonists that have addressed the limitations in current cancer immunotherapy. Immunomodulation of TLR agonists can be classified into two different approaches: (1) molecular modulation via chemical structure modification and (2) macroscopic modulation via an engineered drug delivery system. In molecular modulation, based on prodrug and antedrug principles, activity is modulated (active or inactive) through immolative chemical linkers that can respond to extrinsic or intrinsic biological stimulation and the plasmatic environment, respectively. To increase the effectiveness of TLR agonists as immunostimulatory agents, researchers have conjugated TLR agonists with other immunotherapeutic moieties (antigen, antibody, other TLR agonist, etc.). For macroscopic modulation, bioengineering of delivery carriers differing in size or with albumin hitchhiking moieties has been utilized to increase the efficiency of the targeting of these carriers to secondary lymphoid organs (lymph nodes (LNs) and spleen). The conjugation of specific targeting ligands and incorporation of stimulus-triggering moieties can promote the delivery of TLR agonists into specific cells or intracellular compartments. Implantable porous scaffolds for specific immune cell recruitment and in situ depot-forming gel systems for controlled release of immunomodulatory drugs can increase the therapeutic efficacy of TLR agonists while reducing systemic toxicity. Taken together, these findings show that well-designed and precisely controlled chemical strategies for the immunomodulation of TLR agonists at both the molecular and macroscopic levels are expected to play key roles in improving the therapeutic efficacy of cancer immunotherapy while minimizing immune-related toxicity.
Collapse
Affiliation(s)
- Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, Biomedical Institute for Convergence at SKKU and School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| |
Collapse
|
38
|
Corder BN, Bullard BL, Poland GA, Weaver EA. A Decade in Review: A Systematic Review of Universal Influenza Vaccines in Clinical Trials during the 2010 Decade. Viruses 2020; 12:E1186. [PMID: 33092070 PMCID: PMC7589362 DOI: 10.3390/v12101186] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/31/2022] Open
Abstract
On average, there are 3-5 million severe cases of influenza virus infections globally each year. Seasonal influenza vaccines provide limited protection against divergent influenza strains. Therefore, the development of a universal influenza vaccine is a top priority for the NIH. Here, we report a comprehensive summary of all universal influenza vaccines that were tested in clinical trials during the 2010-2019 decade. Of the 1597 studies found, 69 eligible clinical trials, which investigated 27 vaccines, were included in this review. Information from each trial was compiled for vaccine target, vaccine platform, adjuvant inclusion, clinical trial phase, and results. As we look forward, there are currently three vaccines in phase III clinical trials which could provide significant improvement over seasonal influenza vaccines. This systematic review of universal influenza vaccine clinical trials during the 2010-2019 decade provides an update on the progress towards an improved influenza vaccine.
Collapse
Affiliation(s)
- Brigette N. Corder
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, 4240 Fair Street, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
| | - Brianna L. Bullard
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, 4240 Fair Street, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
| | - Gregory A. Poland
- Mayo Vaccine Research Group, General Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA;
| | - Eric A. Weaver
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, 4240 Fair Street, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
| |
Collapse
|
39
|
Brisse M, Vrba SM, Kirk N, Liang Y, Ly H. Emerging Concepts and Technologies in Vaccine Development. Front Immunol 2020; 11:583077. [PMID: 33101309 PMCID: PMC7554600 DOI: 10.3389/fimmu.2020.583077] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/14/2020] [Indexed: 01/05/2023] Open
Abstract
Despite the success of vaccination to greatly mitigate or eliminate threat of diseases caused by pathogens, there are still known diseases and emerging pathogens for which the development of successful vaccines against them is inherently difficult. In addition, vaccine development for people with compromised immunity and other pre-existing medical conditions has remained a major challenge. Besides the traditional inactivated or live attenuated, virus-vectored and subunit vaccines, emerging non-viral vaccine technologies, such as viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer innovative approaches to address existing challenges of vaccine development. They have also significantly advanced our understanding of vaccine immunology and can guide future vaccine development for many diseases, including rapidly emerging infectious diseases, such as COVID-19, and diseases that have not traditionally been addressed by vaccination, such as cancers and substance abuse. This review provides an integrative discussion of new non-viral vaccine development technologies and their use to address the most fundamental and ongoing challenges of vaccine development.
Collapse
Affiliation(s)
- Morgan Brisse
- Biochemistry, Molecular Biology, and Biophysics Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Natalie Kirk
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
- Comparative Molecular Biosciences Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| |
Collapse
|
40
|
Moser BA, Escalante-Buendia Y, Steinhardt RC, Rosenberger MG, Cassaidy BJ, Naorem N, Chon AC, Nguyen MH, Tran NT, Esser-Kahn AP. Small Molecule NF-κB Inhibitors as Immune Potentiators for Enhancement of Vaccine Adjuvants. Front Immunol 2020; 11:511513. [PMID: 33072085 PMCID: PMC7544742 DOI: 10.3389/fimmu.2020.511513] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 08/25/2020] [Indexed: 12/22/2022] Open
Abstract
Adjuvants are added to vaccines to enhance the immune response and provide increased protection against disease. In the last decade, hundreds of synthetic immune adjuvants have been created, but many induce undesirable levels of proinflammatory cytokines including TNF-α and IL-6. Here we present small molecule NF-κB inhibitors that can be used in combination with an immune adjuvant to both decrease markers associated with poor tolerability and improve the protective response of vaccination. Additionally, we synthesize a library of honokiol derivatives identifying several promising candidates for use in vaccine formulations.
Collapse
Affiliation(s)
- Brittany A Moser
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | | | - Rachel C Steinhardt
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Matthew G Rosenberger
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Britteny J Cassaidy
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Nihesh Naorem
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Alfred C Chon
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Minh H Nguyen
- Department of Chemistry, Chemical Engineering and Materials Science, Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Ngoctran T Tran
- Department of Chemistry, Chemical Engineering and Materials Science, Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Aaron P Esser-Kahn
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
41
|
Challenges for the Newborn Immune Response to Respiratory Virus Infection and Vaccination. Vaccines (Basel) 2020; 8:vaccines8040558. [PMID: 32987691 PMCID: PMC7712002 DOI: 10.3390/vaccines8040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
The initial months of life reflect an extremely challenging time for newborns as a naïve immune system is bombarded with a large array of pathogens, commensals, and other foreign entities. In many instances, the immune response of young infants is dampened or altered, resulting in increased susceptibility and disease following infection. This is the result of both qualitative and quantitative changes in the response of multiple cell types across the immune system. Here we provide a review of the challenges associated with the newborn response to respiratory viral pathogens as well as the hurdles and advances for vaccine-mediated protection.
Collapse
|
42
|
Shang T, Yu X, Han S, Yang B. Nanomedicine-based tumor photothermal therapy synergized immunotherapy. Biomater Sci 2020; 8:5241-5259. [PMID: 32996922 DOI: 10.1039/d0bm01158d] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The emerging anti-tumor immunotherapy has made significant progress in clinical application. However, single immunotherapy is not effective for all anti-tumor treatments, owing to the low objective response rate and the risk of immune-related side effects. Meanwhile, photothermal therapy (PTT) has attracted significant attention because of its non-invasiveness, spatiotemporal controllability and small side effects. Combining PTT with immunotherapy overcomes the issue that single photothermal therapy cannot eradicate tumors with metastasis and recurrence. However, it improves the therapeutic effect of immunotherapy, as the photothermal therapy usually promotes release of tumor-related antigens, triggers immune response by the immunogenic cell death (ICD), thereby, endowing unique synergistic mechanisms for cancer therapy. This review summarizes recent research advances in utilizing nanomedicines for PTT in combination with immunotherapy to improve the outcome of cancer treatment. The strategies include immunogenic cell death, immune agonists and cancer vaccines, immune checkpoint blockades and tumor specific monoclonal antibodies, and small-molecule immune inhibitors. The combination of synergized PTT-immunotherapy with other therapeutic strategies is also discussed.
Collapse
Affiliation(s)
- Tongyi Shang
- The Sixth Affiliated Hospital, Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | | | | | | |
Collapse
|
43
|
Moser BA, Steinhardt RC, Escalante-Buendia Y, Boltz DA, Barker KM, Cassaidy BJ, Rosenberger MG, Yoo S, McGonnigal BG, Esser-Kahn AP. Increased vaccine tolerability and protection via NF-κB modulation. SCIENCE ADVANCES 2020; 6:eaaz8700. [PMID: 32917696 PMCID: PMC11206472 DOI: 10.1126/sciadv.aaz8700] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 07/24/2020] [Indexed: 05/20/2023]
Abstract
Improving adjuvant responses is a promising pathway to develop vaccines against some pathogens (e.g., HIV or dengue). One challenge in adjuvant development is modulating the inflammatory response, which can cause excess side effects, while maintaining immune activation and protection. No approved adjuvants yet have the capability to independently modulate inflammation and protection. Here, we demonstrate a method to limit inflammation while retaining and often increasing the protective responses. To accomplish this goal, we combined a partial selective nuclear factor kappa B (NF-kB) inhibitor with several current adjuvants. The resulting vaccines reduce systemic inflammation and boost protective responses. In an influenza challenge model, we demonstrate that this approach enhances protection. This method was tested across a broad range of adjuvants and antigens. We anticipate these studies will lead to an alternative approach to vaccine formulation design that may prove broadly applicable to a wide range of adjuvants and vaccines.
Collapse
Affiliation(s)
- B A Moser
- Pritzker School for Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA
| | - R C Steinhardt
- Pritzker School for Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA
| | - Y Escalante-Buendia
- Pritzker School for Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA
| | - D A Boltz
- Division of Microbiology and Molecular Biology, IIT Research Institute, Illinois Institute of Technology, 10W. 35th Street, Chicago, IL 60616, USA
| | - K M Barker
- Division of Microbiology and Molecular Biology, IIT Research Institute, Illinois Institute of Technology, 10W. 35th Street, Chicago, IL 60616, USA
| | - B J Cassaidy
- Pritzker School for Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA
| | - M G Rosenberger
- Pritzker School for Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA
| | - S Yoo
- Department of Chemistry, Chemical Engineering & Materials Science, Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - B G McGonnigal
- Department of Chemistry, Chemical Engineering & Materials Science, Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - A P Esser-Kahn
- Pritzker School for Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA.
| |
Collapse
|
44
|
Korgaonkar AA, Nguyen S, Li Y, Sekhar D, Subramanian D, Guevarra J, Pang KCH, Santhakumar V. Distinct cellular mediators drive the Janus faces of toll-like receptor 4 regulation of network excitability which impacts working memory performance after brain injury. Brain Behav Immun 2020; 88:381-395. [PMID: 32259563 PMCID: PMC7415537 DOI: 10.1016/j.bbi.2020.03.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 01/15/2023] Open
Abstract
The mechanisms by which the neurophysiological and inflammatory responses to brain injury contribute to memory impairments are not fully understood. Recently, we reported that the innate immune receptor, toll-like receptor 4 (TLR4) enhances AMPA receptor (AMPAR) currents and excitability in the dentate gyrus after fluid percussion brain injury (FPI) while limiting excitability in controls. Here, we examine the cellular mediators underlying TLR4 regulation of dentate excitability and its impact on memory performance. In ex vivo slices, astrocytic and microglial metabolic inhibitors selectively abolished TLR4 antagonist modulation of excitability in controls, but not in rats after FPI, demonstrating that glial signaling contributes to TLR4 regulation of excitability in controls. In glia-depleted neuronal cultures from naïve mice, TLR4 ligands bidirectionally modulated AMPAR charge transfer consistent with neuronal TLR4 regulation of excitability, as observed after brain injury. In vivo TLR4 antagonism reduced early post-injury increases in mediators of MyD88-dependent and independent TLR4 signaling without altering expression in controls. Blocking TNFα, a downstream effector of TLR4, mimicked effects of TLR4 antagonist and occluded TLR4 agonist modulation of excitability in slices from both control and FPI rats. Functionally, transiently blocking TLR4 in vivo improved impairments in working memory observed one week and one month after FPI, while the same treatment impaired memory function in uninjured controls. Together these data identify that distinct cellular signaling mechanisms converge on TNFα to mediate TLR4 modulation of network excitability in the uninjured and injured brain and demonstrate a role for TLR4 in regulation of working memory function.
Collapse
Affiliation(s)
- Akshata A. Korgaonkar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Correspondence: Akshata Korgaonkar, PhD, Department of Neurology, Washington University School of Medicine, 660 South Euclid Ave, Campus box 8111, St Louis, MO 63110, Phone (Off): 314.362.2999,
| | - Susan Nguyen
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Ying Li
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Dipika Sekhar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Deepak Subramanian
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Jenieve Guevarra
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Kevin C H Pang
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Neurobehavioral Research Lab, Department of Veteran Affairs Medical Center–New Jersey Health Care System, East Orange, New Jersey
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| |
Collapse
|
45
|
Vaccine Design from the Ensemble of Surface Glycoprotein Epitopes of SARS-CoV-2: An Immunoinformatics Approach. Vaccines (Basel) 2020; 8:vaccines8030423. [PMID: 32731461 PMCID: PMC7565012 DOI: 10.3390/vaccines8030423] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/09/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to work out a peptide-based multi-epitope vaccine against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We predicted different B-cell and T-cell epitopes by using the Immune Epitopes Database (IEDB). Homology modeling of the construct was done using SWISS-MODEL and then docked with different toll-like-receptors (TLR4, TLR7, and TLR8) using PatchDock, HADDOCK, and FireDock, respectively. From the overlapped epitopes, we designed five vaccine constructs C1–C5. Based on antigenicity, allergenicity, solubility, different physiochemical properties, and molecular docking scores, we selected the vaccine construct 1 (C1) for further processing. Docking of C1 with TLR4, TLR7, and TLR8 showed striking interactions with global binding energy of −43.48, −65.88, and −60.24 Kcal/mol, respectively. The docked complex was further simulated, which revealed that both molecules remain stable with minimum RMSF. Activation of TLRs induces downstream pathways to produce pro-inflammatory cytokines against viruses and immune system simulation shows enhanced antibody production after the booster dose. In conclusion, C1 was the best vaccine candidate among all designed constructs to elicit an immune response SARS-CoV-2 and combat the coronavirus disease (COVID-19).
Collapse
|
46
|
Harnessing the Complete Repertoire of Conventional Dendritic Cell Functions for Cancer Immunotherapy. Pharmaceutics 2020; 12:pharmaceutics12070663. [PMID: 32674488 PMCID: PMC7408110 DOI: 10.3390/pharmaceutics12070663] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023] Open
Abstract
The onset of checkpoint inhibition revolutionized the treatment of cancer. However, studies from the last decade suggested that the sole enhancement of T cell functionality might not suffice to fight malignancies in all individuals. Dendritic cells (DCs) are not only part of the innate immune system, but also generals of adaptive immunity and they orchestrate the de novo induction of tolerogenic and immunogenic T cell responses. Thus, combinatorial approaches addressing DCs and T cells in parallel represent an attractive strategy to achieve higher response rates across patients. However, this requires profound knowledge about the dynamic interplay of DCs, T cells, other immune and tumor cells. Here, we summarize the DC subsets present in mice and men and highlight conserved and divergent characteristics between different subsets and species. Thereby, we supply a resource of the molecular players involved in key functional features of DCs ranging from their sentinel function, the translation of the sensed environment at the DC:T cell interface to the resulting specialized T cell effector modules, as well as the influence of the tumor microenvironment on the DC function. As of today, mostly monocyte derived dendritic cells (moDCs) are used in autologous cell therapies after tumor antigen loading. While showing encouraging results in a fraction of patients, the overall clinical response rate is still not optimal. By disentangling the general aspects of DC biology, we provide rationales for the design of next generation DC vaccines enabling to exploit and manipulate the described pathways for the purpose of cancer immunotherapy in vivo. Finally, we discuss how DC-based vaccines might synergize with checkpoint inhibition in the treatment of malignant diseases.
Collapse
|
47
|
OMV Vaccines and the Role of TLR Agonists in Immune Response. Int J Mol Sci 2020; 21:ijms21124416. [PMID: 32575921 PMCID: PMC7352230 DOI: 10.3390/ijms21124416] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/26/2022] Open
Abstract
Outer Membrane Vesicles (OMVs) are bacterial nanoparticles that are spontaneously released during growth both in vitro and in vivo by Gram-negative bacteria. They are spherical, bilayered membrane nanostructures that contain many components found within the external surface of the parent bacterium. Naturally, OMVs serve the bacteria as a mechanism to deliver DNA, RNA, proteins, and toxins, as well as to promote biofilm formation and remodel the outer membrane during growth. On the other hand, as OMVs possess the optimal size to be uptaken by immune cells, and present a range of surface-exposed antigens in native conformation and Toll-like receptor (TLR) activating components, they represent an attractive and powerful vaccine platform able to induce both humoral and cell-mediated immune responses. This work reviews the TLR-agonists expressed on OMVs and their capability to trigger individual TLRs expressed on different cell types of the immune system, and then focuses on their impact on the immune responses elicited by OMVs compared to traditional vaccines.
Collapse
|
48
|
Lu H, Zhang X, Wang Y, Zong Y, Wang Y, Zhang X, Xia X, Sun H. Superior adjuvanticity of the genetically fused D1 domain of Neisseria meningitides Ag473 lipoprotein among three Toll-like receptor ligands. Biosci Rep 2020; 40:BSR20193675. [PMID: 32202301 PMCID: PMC7138904 DOI: 10.1042/bsr20193675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/04/2020] [Accepted: 03/17/2020] [Indexed: 11/17/2022] Open
Abstract
Toll-like receptor (TLR) ligands have emerged as the attractive adjuvant for subunit vaccines. However, selection of TLR ligands needs to be rationally chosen on the basis of antigen and adjuvant properties. In the present study, we expressed the Ag473 lipoprotein from Neisseria meningitides, flagellin FlaB from Vibrio vulnificus and heat shock protein 70 from Mycobacterium tuberculosis (mHsp70) in Escherichia coli as single proteins and fusion proteins with VP2 protein of infectious bursal disease virus (IBDV). Both cellular and humoral adjuvanticities of the three TLR ligands were compared by immunization of mice in two different ways. Among the three co-administered TLR ligands, recombinant Ag473 lipoprotein exhibited the highest cellular and humoral adjuvanticities, including promotion of IL-4, IL-12, IFN-γ and IBDV VP2-specific antibody production. Among the three genetically fused TLR ligands, fusion with Ag473 D1 domain exhibited the highest cellular and humoral adjuvanticities. Overall, the adjuvanticities of genetically fused TRL ligands were significantly higher than that of co-administered TLR ligands. Fusion with Ag473 D1 domain exhibited superior adjuvanticity among the three TLR ligands delivered in two different ways.
Collapse
Affiliation(s)
- Huipeng Lu
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiaokai Zhang
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Yuyang Wang
- The Testing Center, Yangzhou University, Yangzhou 225009, China
| | - Yang Zong
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Yajie Wang
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xinyu Zhang
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiaoli Xia
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Huaichang Sun
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China
| |
Collapse
|
49
|
Han Y, Zhu L, Wu W, Zhang H, Hu W, Dai L, Yang Y. Small Molecular Immune Modulators as Anticancer Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:547-618. [PMID: 32185725 DOI: 10.1007/978-981-15-3266-5_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
After decades of intense effort, immune checkpoint inhibitors have been conclusively demonstrated to be effective in cancer treatments and thus are revolutionizing the concepts in the treatment of cancers. Immuno-oncology has arrived and will play a key role in cancer treatment in the foreseeable future. However, efforts to find novel methods to improve the immune response to cancer have not ceased. Small-molecule approaches offer inherent advantages over biologic immunotherapies since they can cross cell membranes, penetrate into tumor tissue and tumor microenvironment more easily, and are amenable to be finely controlled than biological agents, which may help reduce immune-related adverse events seen with biologic therapies and provide more flexibility for the combination use with other therapies and superior clinical benefit. On the one hand, small-molecule therapies can modulate the immune response to cancer by restoring the antitumor immunity, promoting more effective cytotoxic lymphocyte responses, and regulating tumor microenvironment, either directly or epigenetically. On the other hand, the combination of different mechanisms of small molecules with antibodies and other biologics demonstrated admirable synergistic effect in clinical settings for cancer treatment and may expand antibodies' usefulness for broader clinical applications. This chapter provides an overview of small-molecule immunotherapeutic approaches either as monotherapy or in combination for the treatment of cancer.
Collapse
Affiliation(s)
- Yongxin Han
- Lapam Capital LLC., 17C1, Tower 2, Xizhimenwai Street, Xicheng District, Beijing, 100044, China.
| | - Li Zhu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Wei Wu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Hui Zhang
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Wei Hu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Liguang Dai
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Yanqing Yang
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| |
Collapse
|
50
|
Structural basis and designing of peptide vaccine using PE-PGRS family protein of Mycobacterium ulcerans—An integrated vaccinomics approach. Mol Immunol 2020; 120:146-163. [DOI: 10.1016/j.molimm.2020.02.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/29/2022]
|