1
|
Giram P, Md Mahabubur Rahman K, Aqel O, You Y. In Situ Cancer Vaccines: Redefining Immune Activation in the Tumor Microenvironment. ACS Biomater Sci Eng 2025. [PMID: 40223683 DOI: 10.1021/acsbiomaterials.5c00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Cancer is one of the leading causes of mortality worldwide. Nanomedicines have significantly improved life expectancy and survival rates for cancer patients in current standard care. However, recurrence of cancer due to metastasis remains a significant challenge. Vaccines can provide long-term protection and are ideal for preventing bacterial and viral infections. Cancer vaccines, however, have shown limited therapeutic efficacy and raised safety concerns despite extensive research. Cancer vaccines target and stimulate responses against tumor-specific antigens and have demonstrated great potential for cancer treatment in preclinical studies. However, tumor-associated immunosuppression and immune tolerance driven by immunoediting pose significant challenges for vaccine design. In situ vaccination represents an alternative approach to traditional cancer vaccines. This strategy involves the intratumoral administration of immunostimulants to modulate the growth and differentiation of innate immune cells, such as dendritic cells, macrophages, and neutrophils, and restore T-cell activity. Currently approved in situ vaccines, such as T-VEC, have demonstrated clinical promise, while ongoing clinical trials continue to explore novel strategies for broader efficacy. Despite these advancements, failures in vaccine research highlight the need to address tumor-associated immune suppression and immune escape mechanisms. In situ vaccination strategies combine innate and adaptive immune stimulation, leveraging tumor-associated antigens to activate dendritic cells and cross-prime CD8+ T cells. Various vaccine modalities, such as nucleotide-based vaccines (e.g., RNA and DNA vaccines), peptide-based vaccines, and cell-based vaccines (including dendritic, T-cell, and B-cell approaches), show significant potential. Plant-based viral approaches, including cowpea mosaic virus and Newcastle disease virus, further expand the toolkit for in situ vaccination. Therapeutic modalities such as chemotherapy, radiation, photodynamic therapy, photothermal therapy, and Checkpoint blockade inhibitors contribute to enhanced antigen presentation and immune activation. Adjuvants like CpG-ODN and PRR agonists further enhance immune modulation and vaccine efficacy. The advantages of in situ vaccination include patient specificity, personalization, minimized antigen immune escape, and reduced logistical costs. However, significant barriers such as tumor heterogeneity, immune evasion, and logistical challenges remain. This review explores strategies for developing potent cancer vaccines, examines ongoing clinical trials, evaluates immune stimulation methods, and discusses prospects for advancing in situ cancer vaccination.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Osama Aqel
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
2
|
Delgado-Almenta V, Blaya-Cánovas JL, Calahorra J, López-Tejada A, Griñán-Lisón C, Granados-Principal S. Cancer Vaccines and Beyond: The Transformative Role of Nanotechnology in Immunotherapy. Pharmaceutics 2025; 17:216. [PMID: 40006583 PMCID: PMC11859086 DOI: 10.3390/pharmaceutics17020216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality globally, responsible for approximately 10 million deaths in 2022 and an estimated 21 million new cases in 2024. Traditional cancer treatments such as surgery, radiation therapy, and chemotherapy often present limitations in efficacy and side effects. However, immunotherapeutic vaccines have emerged as a promising approach, leveraging the body's immune system to target and eliminate cancer cells. This review examines the evolving landscape of cancer vaccines, differentiating between preventive and therapeutic strategies and highlighting the significance of tumor-specific antigens, including tumor-associated antigens (TAAs) and neoantigens. Recent advancements in vaccine technology, particularly through nanotechnology, have resulted in the development of nanovaccines, which enhance antigen stability, optimize delivery to immune cells, and promote robust immune responses. Notably, clinical data indicate that patients receiving immune checkpoint inhibitors can achieve overall survival rates of approximately 34.8 months compared to just 15.7 months for traditional therapies. Despite these advancements, challenges remain, such as the immunosuppressive tumor microenvironment and tumor heterogeneity. Emerging evidence suggests that combining nanovaccines with immunomodulators may enhance therapeutic efficacy by overcoming these obstacles. Continued research and interdisciplinary collaboration will be essential to fully exploit the promise of nanovaccines, ultimately leading to more effective and accessible treatments for cancer patients. The future of cancer immunotherapy appears increasingly hopeful as these innovative strategies pave the way for enhanced patient outcomes and an improved quality of life in oncology.
Collapse
Affiliation(s)
- Violeta Delgado-Almenta
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Jesús Calahorra
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| | - Carmen Griñán-Lisón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), Centro de Investigación Biomédica (CIBM), University of Granada, 18016 Granada, Spain
| | - Sergio Granados-Principal
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| |
Collapse
|
3
|
Zhou W, Lu X, Tian F, Luo Q, Zhou W, Yang S, Li W, Yang Y, Shi M, Zhou T. Vaccine Therapies for Prostate Cancer: Current Status and Future Outlook. Vaccines (Basel) 2024; 12:1384. [PMID: 39772046 PMCID: PMC11679746 DOI: 10.3390/vaccines12121384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Prostate cancer is a prevalent cancer in elderly men, and immunotherapy has emerged as a promising treatment approach in recent years. The aim of immunotherapy is to stimulate the body's immune system to target and destroy cancer cells. Cancer vaccines that are highly specific, safe, and capable of creating long-lasting immune responses are a key focus in cancer immunotherapy research. Despite progress in clinical trials showing positive results, the practical use of cancer vaccines still encounters various obstacles. The complexity of the immune microenvironment and variations in the immune systems of individual patients have hindered the progress of research on prostate cancer vaccines. This review examines the history and mechanisms of cancer vaccines, summarizes recent clinical research findings, and explores future directions in the development of prostate cancer vaccines.
Collapse
Affiliation(s)
- Wenhao Zhou
- Department of Urology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China; (W.Z.); (X.L.)
| | - Xiaojun Lu
- Department of Urology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China; (W.Z.); (X.L.)
| | - Feng Tian
- Department of Urology, Shanghai Eighth People’s Hospital, Shanghai 200235, China;
| | - Qianming Luo
- School of Medicine, Tongji University, Shanghai 200092, China; (Q.L.); (W.Z.); (S.Y.)
| | - Weihang Zhou
- School of Medicine, Tongji University, Shanghai 200092, China; (Q.L.); (W.Z.); (S.Y.)
| | - Siyuan Yang
- School of Medicine, Tongji University, Shanghai 200092, China; (Q.L.); (W.Z.); (S.Y.)
| | - Wenxuan Li
- College of Clinical Medicine, Naval Medical University, Shanghai 200433, China; (W.L.); (Y.Y.)
| | - Yongjun Yang
- College of Clinical Medicine, Naval Medical University, Shanghai 200433, China; (W.L.); (Y.Y.)
| | - Minfeng Shi
- Reproduction Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tie Zhou
- Department of Urology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China; (W.Z.); (X.L.)
| |
Collapse
|
4
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [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: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
5
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
6
|
Liu D, Liu L, Li X, Wang S, Wu G, Che X. Advancements and Challenges in Peptide-Based Cancer Vaccination: A Multidisciplinary Perspective. Vaccines (Basel) 2024; 12:950. [PMID: 39204073 PMCID: PMC11359700 DOI: 10.3390/vaccines12080950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
With the continuous advancements in tumor immunotherapy, researchers are actively exploring new treatment methods. Peptide therapeutic cancer vaccines have garnered significant attention for their potential in improving patient outcomes. Despite its potential, only a single peptide-based cancer vaccine has been approved by the U.S. Food and Drug Administration (FDA). A comprehensive understanding of the underlying mechanisms and current development status is crucial for advancing these vaccines. This review provides an in-depth analysis of the production principles and therapeutic mechanisms of peptide-based cancer vaccines, highlights the commonly used peptide-based cancer vaccines, and examines the synergistic effects of combining these vaccines with immunotherapy, targeted therapy, radiotherapy, and chemotherapy. While some studies have yielded suboptimal results, the potential of combination therapies remains substantial. Additionally, we addressed the management and adverse events associated with peptide-based cancer vaccines, noting their relatively higher safety profile compared to traditional radiotherapy and chemotherapy. Lastly, we also discussed the roles of adjuvants and targeted delivery systems in enhancing vaccine efficacy. In conclusion, this review comprehensively outlines the current landscape of peptide-based cancer vaccination and underscores its potential as a pivotal immunotherapy approach.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Xinghan Li
- Department of Stomatology, General Hospital of Northern Theater Command, Shenyang 110016, China;
| | - Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| |
Collapse
|
7
|
Wilks LR, Joshi G, Rychener N, Gill HS. Generation of Broad Protection against Influenza with Di-Tyrosine-Cross-Linked M2e Nanoclusters. ACS Infect Dis 2024; 10:1552-1560. [PMID: 38623820 DOI: 10.1021/acsinfecdis.3c00429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.
Collapse
Affiliation(s)
- Logan R Wilks
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Natalie Rychener
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
8
|
Dehghankhold M, Sadat Abolmaali S, Nezafat N, Mohammad Tamaddon A. Peptide nanovaccine in melanoma immunotherapy. Int Immunopharmacol 2024; 129:111543. [PMID: 38301413 DOI: 10.1016/j.intimp.2024.111543] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/03/2024]
Abstract
Melanoma is an especially fatal neoplasm resistant to traditional treatment. The advancement of novel therapeutical approaches has gained attention in recent years by shedding light on the molecular mechanisms of melanoma tumorigenesis and their powerful interplay with the immune system. The presence of many mutations in melanoma cells results in the production of a varied array of antigens. These antigens can be recognized by the immune system, thereby enabling it to distinguish between tumors and healthy cells. In the context of peptide cancer vaccines, generally, they are designed based on tumor antigens that stimulate immunity through antigen-presenting cells (APCs). As naked peptides often have low potential in eliciting a desirable immune reaction, immunization with such compounds usually necessitates adjuvants and nanocarriers. Actually, nanoparticles (NPs) can provide a robust immune response to peptide-based melanoma vaccines. They improve the directing of peptide vaccines to APCs and induce the secretion of cytokines to get maximum immune response. This review provides an overview of the current knowledge of the utilization of nanotechnology in peptide vaccines emphasizing melanoma, as well as highlights the significance of physicochemical properties in determining the fate of these nanovaccines in vivo, including their drainage to lymph nodes, cellular uptake, and influence on immune responses.
Collapse
Affiliation(s)
- Mahvash Dehghankhold
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Computational vaccine and Drug Design Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Xiong Z, Raphael I, Olin M, Okada H, Li X, Kohanbash G. Glioblastoma vaccines: past, present, and opportunities. EBioMedicine 2024; 100:104963. [PMID: 38183840 PMCID: PMC10808938 DOI: 10.1016/j.ebiom.2023.104963] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/08/2024] Open
Abstract
Glioblastoma (GBM) is one of the most lethal central nervous systems (CNS) tumours in adults. As supplements to standard of care (SOC), various immunotherapies improve the therapeutic effect in other cancers. Among them, tumour vaccines can serve as complementary monotherapy or boost the clinical efficacy with other immunotherapies, such as immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapy. Previous studies in GBM therapeutic vaccines have suggested that few neoantigens could be targeted in GBM due to low mutation burden, and single-peptide therapeutic vaccination had limited efficacy in tumour control as monotherapy. Combining diverse antigens, including neoantigens, tumour-associated antigens (TAAs), and pathogen-derived antigens, and optimizing vaccine design or vaccination strategy may help with clinical efficacy improvement. In this review, we discussed current GBM therapeutic vaccine platforms, evaluated and potential antigenic targets, current challenges, and perspective opportunities for efficacy improvement.
Collapse
Affiliation(s)
- Zujian Xiong
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA; Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Michael Olin
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008 PR China.
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
10
|
Zahedipour F, Jamialahmadi K, Zamani P, Reza Jaafari M. Improving the efficacy of peptide vaccines in cancer immunotherapy. Int Immunopharmacol 2023; 123:110721. [PMID: 37543011 DOI: 10.1016/j.intimp.2023.110721] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Peptide vaccines have shown great potential in cancer immunotherapy by targeting tumor antigens and activating the patient's immune system to mount a specific response against cancer cells. However, the efficacy of peptide vaccines in inducing a sustained immune response and achieving clinical benefit remains a major challenge. In this review, we discuss the current status of peptide vaccines in cancer immunotherapy and strategies to improve their efficacy. We summarize the recent advancements in the development of peptide vaccines in pre-clinical and clinical settings, including the use of novel adjuvants, neoantigens, nano-delivery systems, and combination therapies. We also highlight the importance of personalized cancer vaccines, which consider the unique genetic and immunological profiles of individual patients. We also discuss the strategies to enhance the immunogenicity of peptide vaccines such as multivalent peptides, conjugated peptides, fusion proteins, and self-assembled peptides. Although, peptide vaccines alone are weak immunogens, combining peptide vaccines with other immunotherapeutic approaches and developing novel approaches such as personalized vaccines can be promising methods to significantly enhance their efficacy and improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Guo Y, Gao F, Ahmed A, Rafiq M, Yu B, Cong H, Shen Y. Immunotherapy: cancer immunotherapy and its combination with nanomaterials and other therapies. J Mater Chem B 2023; 11:8586-8604. [PMID: 37614168 DOI: 10.1039/d3tb01358h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Immunotherapy is a new type of tumor treatment after surgery, radiotherapy and chemotherapy, and can be used to manage and destroy tumor cells through activating or strengthening the immune response. Immunotherapy has the benefits of a low recurrence rate and high specificity compared to traditional treatment methods. Immunotherapy has developed rapidly in recent years and has become a research hotspot. Currently, chimeric antigen receptor T-cell immunotherapy and immune checkpoint inhibitors are the most effective tumor immunotherapies in clinical practice. While tumor immunotherapy brings hope to patients, it also faces some challenges and still requires continuous research and progress. Combination therapy is the future direction of anti-tumor treatment. In this review, the main focus is on an overview of the research progress of immune checkpoint inhibitors, cellular therapies, tumor vaccines, small molecule inhibitors and oncolytic virotherapy in tumor treatment, as well as the combination of immunotherapy with other treatments.
Collapse
Affiliation(s)
- Yuanyuan Guo
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China.
| | - Fengyuan Gao
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China.
| | - Adeel Ahmed
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China.
| | - Muhammad Rafiq
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China.
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China.
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China.
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
- School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China.
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
12
|
Pérez-Baños A, Gleisner MA, Flores I, Pereda C, Navarrete M, Araya JP, Navarro G, Quezada-Monrás C, Tittarelli A, Salazar-Onfray F. Whole tumour cell-based vaccines: tuning the instruments to orchestrate an optimal antitumour immune response. Br J Cancer 2023; 129:572-585. [PMID: 37355722 PMCID: PMC10421921 DOI: 10.1038/s41416-023-02327-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023] Open
Abstract
Immunotherapy, particularly those based on immune checkpoint inhibitors (ICIs), has become a useful approach for many neoplastic diseases. Despite the improvements of ICIs in supporting tumour regression and prolonging survival, many patients do not respond or develop resistance to treatment. Thus, therapies that enhance antitumour immunity, such as anticancer vaccines, constitute a feasible and promising therapeutic strategy. Whole tumour cell (WTC) vaccines have been extensively tested in clinical studies as intact or genetically modified cells or tumour lysates, injected directly or loaded on DCs with distinct adjuvants. The essential requirements of WTC vaccines include the optimal delivery of a broad battery of tumour-associated antigens, the presence of tumour cell-derived molecular danger signals, and adequate adjuvants. These factors trigger an early and robust local innate inflammatory response that orchestrates an antigen-specific and proinflammatory adaptive antitumour response capable of controlling tumour growth by several mechanisms. In this review, the strengths and weaknesses of our own and others' experiences in studying WTC vaccines are revised to discuss the essential elements required to increase anticancer vaccine effectiveness.
Collapse
Affiliation(s)
- Amarilis Pérez-Baños
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Iván Flores
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mariela Navarrete
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Juan Pablo Araya
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Giovanna Navarro
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, 5110566, Chile
| | - Claudia Quezada-Monrás
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, 5110566, Chile
| | - Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación (PIDi), Universidad Tecnológica Metropolitana (UTEM), Santiago, Chile.
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute and Section for Infectious Diseases, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
13
|
Matos AS, Invenção MDCV, Moura IAD, Freitas ACD, Batista MVDA. Immunoinformatics applications in the development of therapeutic vaccines against human papillomavirus-related infections and cervical cancer. Rev Med Virol 2023; 33:e2463. [PMID: 37291746 DOI: 10.1002/rmv.2463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/09/2023] [Accepted: 05/27/2023] [Indexed: 06/10/2023]
Abstract
The human papillomavirus (HPV) represents the most prevalent sexually transmitted infectious agent worldwide. HPV penetrates the epithelium through microlesions and establishes an infectious focus that can lead to the development of cervical cancer. Prophylactic HPV vaccines are available, but do not affect already-established infections. Using in silico prediction tools is a promising strategy for identifying and selecting vaccine candidate T cell epitopes. An advantage of this strategy is that epitopes can be selected according to the degree of conservation within a group of antigenic proteins. This makes achieving comprehensive genotypic coverage possible with a small set of epitopes. Therefore, this paper revises the general characteristics of HPV biology and the current knowledge on developing therapeutic peptide vaccines against HPV-related infections and cervical cancer.
Collapse
Affiliation(s)
- Alexandre Santos Matos
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, Sao Cristovao, Brazil
| | - Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Federal University of Pernambuco, Recife, Brazil
| | - Ingrid Andrêssa de Moura
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Federal University of Pernambuco, Recife, Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Federal University of Pernambuco, Recife, Brazil
| | - Marcus Vinicius de Aragão Batista
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, Sao Cristovao, Brazil
| |
Collapse
|
14
|
Hou Y, Chen M, Bian Y, Zheng X, Tong R, Sun X. Advanced subunit vaccine delivery technologies: From vaccine cascade obstacles to design strategies. Acta Pharm Sin B 2023; 13:3321-3338. [PMID: 37655334 PMCID: PMC10465871 DOI: 10.1016/j.apsb.2023.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Designing and manufacturing safe and effective vaccines is a crucial challenge for human health worldwide. Research on adjuvant-based subunit vaccines is increasingly being explored to meet clinical needs. Nevertheless, the adaptive immune responses of subunit vaccines are still unfavorable, which may partially be attributed to the immune cascade obstacles and unsatisfactory vaccine design. An extended understanding of the crosstalk between vaccine delivery strategies and immunological mechanisms could provide scientific insight to optimize antigen delivery and improve vaccination efficacy. In this review, we summarized the advanced subunit vaccine delivery technologies from the perspective of vaccine cascade obstacles after administration. The engineered subunit vaccines with lymph node and specific cell targeting ability, antigen cross-presentation, T cell activation properties, and tailorable antigen release patterns may achieve effective immune protection with high precision, efficiency, and stability. We hope this review can provide rational design principles and inspire the exploitation of future subunit vaccines.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xi Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Zhao J, Liao B, Gong L, Yang H, Li S, Li Y. Knowledge mapping of therapeutic cancer vaccine from 2013 to 2022: A bibliometric and visual analysis. Hum Vaccin Immunother 2023; 19:2254262. [PMID: 37728107 PMCID: PMC10512878 DOI: 10.1080/21645515.2023.2254262] [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: 07/12/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
The investigation of therapeutic cancer vaccines has been ongoing for the past century. Herein, we used VOSviewer and CiteSpace to perform the first global bibliometric analysis of the literature on therapeutic cancer vaccines from 2013 to 2022 aiming to explore the current status and potential research trends. The findings revealed a consistent upward trend in both publication counts and citations. The United States emerged as the leading contributor with the highest number of published papers. Additionally, the analysis of references and keywords indicated that therapeutic cancer vaccines have long been popular topics, whereas neoantigen vaccines, mRNA vaccines, combination strategies, and vaccine delivery systems are emerging research hotspots. This bibliometric study provides a comprehensive and important overview of the current knowledge and potential developments in therapeutic cancer vaccines from 2013 to 2022, which may serve as a valuable reference for scholars interested in further exploring this promising field.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Bin Liao
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Li Gong
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Huiyao Yang
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Sha Li
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
16
|
Zhang H, Zhang Y, Hu H, Yang W, Xia X, Lei L, Lin R, Li J, Li Y, Gao H. In Situ Tumor Vaccine for Lymph Nodes Delivery and Cancer Therapy Based on Small Size Nanoadjuvant. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301041. [PMID: 37078903 DOI: 10.1002/smll.202301041] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/26/2023] [Indexed: 05/03/2023]
Abstract
Tumor vaccine is a promising cancer treatment modality, however, the convenient antigens loading in vivo and efficient delivery of vaccines to lymph nodes (LNs) still remain a formidable challenge. Herein, an in situ nanovaccine strategy targeting LNs to induce powerful antitumor immune responses by converting the primary tumor into whole-cell antigens and then delivering these antigens and nanoadjuvants simultaneously to LNs is proposed. The in situ nanovaccine is based on a hydrogel system, which loaded with doxorubicin (DOX) and nanoadjuvant CpG-P-ss-M. The gel system exhibits ROS-responsive release of DOX and CpG-P-ss-M, generating abundant in situ storage of whole-cell tumor antigens. CpG-P-ss-M adsorbs tumor antigens through the positive surface charge and achieves charge reversal, forming small-sized and negatively charged tumor vaccines in situ, which are then primed to LNs. Eventually, the tumor vaccine promotes antigens uptake by dendritic cells (DCs), maturation of DCs, and proliferation of T cells. Moreover, the vaccine combined with anti-CTLA4 antibody and losartan inhibits tumor growth by 50%, significantly increasing the percentage of splenic cytotoxic T cells (CTLs), and generating tumor-specific immune responses. Overall, the treatment effectively inhibits primary tumor growth and induces tumor-specific immune response. This study provides a scalable strategy for in situ tumor vaccination.
Collapse
Affiliation(s)
- Huilin Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yiwei Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Haili Hu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Wenqin Yang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xue Xia
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Lei Lei
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Ruyi Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jiamei Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yuan Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
17
|
Nordin ML, Azemi AK, Nordin AH, Nabgan W, Ng PY, Yusoff K, Abu N, Lim KP, Zakaria ZA, Ismail N, Azmi F. Peptide-Based Vaccine against Breast Cancer: Recent Advances and Prospects. Pharmaceuticals (Basel) 2023; 16:923. [PMID: 37513835 PMCID: PMC10386531 DOI: 10.3390/ph16070923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer is considered the second-leading cancer after lung cancer and is the most prevalent cancer among women globally. Currently, cancer immunotherapy via vaccine has gained great attention due to specific and targeted immune cell activity that creates a potent immune response, thus providing long-lasting protection against the disease. Despite peptides being very susceptible to enzymatic degradation and poor immunogenicity, they can be easily customized with selected epitopes to induce a specific immune response and particulate with carriers to improve their delivery and thus overcome their weaknesses. With advances in nanotechnology, the peptide-based vaccine could incorporate other components, thereby modulating the immune system response against breast cancer. Considering that peptide-based vaccines seem to show remarkably promising outcomes against cancer, this review focuses on and provides a specific view of peptide-based vaccines used against breast cancer. Here, we discuss the benefits associated with a peptide-based vaccine, which can be a mainstay in the prevention and recurrence of breast cancer. Additionally, we also report the results of recent trials as well as plausible prospects for nanotechnology against breast cancer.
Collapse
Affiliation(s)
- Muhammad Luqman Nordin
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia (UKM) Kuala Lumpur Campus, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Malaysia Kelantan (UMK), Pengkalan Chepa, Kota Bharu 16100, Kelantan, Malaysia
| | - Ahmad Khusairi Azemi
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Malaysia
| | - Abu Hassan Nordin
- Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM), Arau 02600, Malaysia
| | - Walid Nabgan
- Departament d'Enginyeria Química, Universitat Rovira I Virgili, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - Pei Yuen Ng
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia (UKM), Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Khatijah Yusoff
- National Institutes of Biotechnology, Malaysia Genome and Vaccine Institute, Jalan Bangi, Kajang 43000, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Centre, Jalan Ya'acob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Kue Peng Lim
- Cancer Immunology & Immunotherapy Unit, Cancer Research Malaysia, No. 1 Jalan SS12/1A, Subang Jaya 47500, Malaysia
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, Kota Kinabalu 88400, Malaysia
| | - Noraznawati Ismail
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Malaysia
| | - Fazren Azmi
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia (UKM) Kuala Lumpur Campus, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
18
|
Zahedipour F, Zamani P, Mashreghi M, Astaneh M, Sankian M, Amiri A, Jamialahmadi K, Jaafari MR. Nanoliposomal VEGF-R2 peptide vaccine acts as an effective therapeutic vaccine in a murine B16F10 model of melanoma. Cancer Nanotechnol 2023; 14:62. [PMID: 37333490 PMCID: PMC10264216 DOI: 10.1186/s12645-023-00213-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
Background The vascular endothelial growth factor receptor-2 (VEGFR-2) plays an important role in melanoma development and progression. Peptide vaccines have shown great potential in cancer immunotherapy by targeting VEGFR-2 as a tumor-associated antigen and boosting the immune response against both tumor cells and tumor endothelial cells. Despite this, the low efficiency of peptide vaccines has resulted in moderate therapeutic results in the majority of studies. Enhancing the delivery of peptide vaccines using nanoliposomes is an important strategy for improving the efficacy of peptide vaccines. In this regard, we designed VEGFR-2-derived peptides restricted to both mouse MHC I and human HLA-A*02:01 using immunoinformatic tools and selected three peptides representing the highest binding affinities. The peptides were encapsulated in nanoliposomal formulations using the film method plus bath sonication and characterized for their colloidal properties. Results The mean diameter of peptide-encapsulated liposomes was around 135 nm, zeta potential of - 17 mV, and encapsulation efficiency of approximately 70%. Then, vaccine formulations were injected subcutaneously in mice bearing B16F10-established melanoma tumors and their efficiency in triggering immunological, and anti-tumor responses was evaluated. Our results represented that one of our designed VEGFR-2 peptide nanoliposomal formulations (Lip-V1) substantially activated CD4+ (p < 0.0001) and CD8+ (P < 0.001) T cell responses and significantly boosted the production of IFN-γ (P < 0.0001) and IL-4 (P < 0.0001). Furthermore, this formulation led to a significant decrease in tumor volume (P < 0.0001) and enhanced survival (P < 0.05) in mice. Conclusion Our findings suggest that the nanoliposomal formulation containing VEGFR-2 peptides could be a promising therapeutic vaccination approach capable of eliciting strong antigen-specific immunologic and anti-tumor responses. Supplementary Information The online version contains supplementary material available at 10.1186/s12645-023-00213-7.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Astaneh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Oladejo M, Paulishak W, Wood L. Synergistic potential of immune checkpoint inhibitors and therapeutic cancer vaccines. Semin Cancer Biol 2023; 88:81-95. [PMID: 36526110 DOI: 10.1016/j.semcancer.2022.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Cancer vaccines and immune checkpoint inhibitors (ICIs) function at different stages of the cancer immune cycle due to their distinct mechanisms of action. Therapeutic cancer vaccines enhance the activation and infiltration of cytotoxic immune cells into the tumor microenvironment (TME), while ICIs, prevent and/or reverse the dysfunction of these immune cells. The efficacy of both classes of immunotherapy has been evaluated in monotherapy, but they have been met with several challenges. Although therapeutic cancer vaccines can activate anti-tumor immune responses, these responses are susceptible to attenuation by immunoregulatory molecules. Similarly, ICIs are ineffective in the absence of tumor-infiltrating lymphocytes (TILs). Further, ICIs are often associated with immune-related adverse effects that may limit quality of life and compliance. However, the combination of the improved immunogenicity afforded by cancer vaccines and restrained immunosuppression provided by immune checkpoint inhibitors may provide a suitable platform for therapeutic synergism. In this review, we revisit the history and various classifications of therapeutic cancer vaccines. We also provide a summary of the currently approved ICIs. Finally, we provide mechanistic insights into the synergism between ICIs and cancer vaccines.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Wyatt Paulishak
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Laurence Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
20
|
Gao C, Chen Y, Cheng X, Zhang Y, Zhang Y, Wang Y, Cui Z, Liao Y, Luo P, Wu W, Wang C, Zeng H, Zou Q, Gu J. A novel structurally identified epitope delivered by macrophage membrane-coated PLGA nanoparticles elicits protection against Pseudomonas aeruginosa. J Nanobiotechnology 2022; 20:532. [PMID: 36517801 PMCID: PMC9750051 DOI: 10.1186/s12951-022-01725-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
The increasing prevalence of antibiotic resistance by Pseudomonas aeruginosa (PA) raises an urgent need for an effective vaccine. The outer membrane proteins of PA, especially those that are upregulated during infection, are ideal vaccine targets. However, the strong hydrophobicity of these proteins hinders their application for this purpose. In this study, we selected eight outer membrane proteins from PA with the most significantly upregulated expression. Their extracellular loops were analyzed and screened by using sera from patients who had recovered from PA infection. As a result, a novel immunogenic epitope (Ep167-193) from PilY1 (PA4554) was found. Moreover, we constructed a macrophage membrane-coated PLGA (poly lactic-co-glycolic acid) nanoparticle vaccine carrying PilY1 Ep167-193 (PNPs@M-Ep167-193) that elicits a Th2 immune response and confers adequate protection in mice. Our data furnished the promising vaccine candidate PNPs@M-Ep167-193 while providing additional evidence for structure-based epitope identification and vaccine design.
Collapse
Affiliation(s)
- Chen Gao
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Yin Chen
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing, Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Xin Cheng
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Yi Zhang
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Yueyue Zhang
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Ying Wang
- grid.410570.70000 0004 1760 6682953Th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, (Third Military Medical University), Shigatse, 857000 China
| | - Zhiyuan Cui
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Yaling Liao
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Ping Luo
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Weihui Wu
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Cheng Wang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Chongqing, Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Hao Zeng
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Quanming Zou
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Jiang Gu
- grid.410570.70000 0004 1760 6682National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, The 30th, Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| |
Collapse
|
21
|
Wilks LR, Joshi G, Kang SM, Wang BZ, Gill HS. Peptide Cross-Linking Using Tyrosine Residues Facilitated by an Exogenous Nickel-Histidine Complex: A Facile Approach for Enhancing Vaccine-Specific Immunogenicity. ACS Infect Dis 2022; 8:2389-2395. [PMID: 36346898 DOI: 10.1021/acsinfecdis.2c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An improved method for the generation of peptide vaccines using di-tyrosine cross-linking is described. The conserved ion channel peptide, M2e, of influenza A virus was modified with the addition of small tyrosine-rich regions (GYGY-) at both the N- and C-termini and extensively cross-linked via tyrosine-tyrosine linkages to form peptide nanoclusters. The cross-linking was catalyzed using exogenous nickel(II) ions complexed to an exogenous glycine-glycine-histidine peptide in the presence of an oxidizer. Mice that were intranasally or intramuscularly immunized with the M2e-vaccine nanoclusters induced comparable levels of M2e-specific serum antibodies. Vaccination via the intranasal or intramuscular route protected mice from subsequent lethal challenge with an influenza A virus. In comparison to our previous approach, where a histidine-rich tag was added into the peptide structure, the use of exogenous histidine reduced irrelevant off-target immune response. Additionally, the purity of the resulting nanoclusters is an attractive feature, making this approach appealing for vaccine development.
Collapse
Affiliation(s)
- Logan R Wilks
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave., Atlanta, Georgia 30302, United States
| | - Bao-Zhong Wang
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave., Atlanta, Georgia 30302, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
22
|
Immunoinformatics Approach for Epitope-Based Vaccine Design: Key Steps for Breast Cancer Vaccine. Diagnostics (Basel) 2022; 12:diagnostics12122981. [PMID: 36552988 PMCID: PMC9777080 DOI: 10.3390/diagnostics12122981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Vaccines are an upcoming medical intervention for breast cancer. By targeting the tumor antigen, cancer vaccines can be designed to train the immune system to recognize tumor cells. Therefore, along with technological advances, the vaccine design process is now starting to be carried out with more rational methods such as designing epitope-based peptide vaccines using immunoinformatics methods. Immunoinformatics methods can assist vaccine design in terms of antigenicity and safety. Common protocols used to design epitope-based peptide vaccines include tumor antigen identification, protein structure analysis, T cell epitope prediction, epitope characterization, and evaluation of protein-epitope interactions. Tumor antigen can be divided into two types: tumor associated antigen and tumor specific antigen. We will discuss the identification of tumor antigens using high-throughput technologies. Protein structure analysis comprises the physiochemical, hydrochemical, and antigenicity of the protein. T cell epitope prediction models are widely available with various prediction parameters as well as filtering tools for the prediction results. Epitope characterization such as allergenicity and toxicity can be done in silico as well using allergenicity and toxicity predictors. Evaluation of protein-epitope interactions can also be carried out in silico with molecular simulation. We will also discuss current and future developments of breast cancer vaccines using an immunoinformatics approach. Finally, although prediction models have high accuracy, the opposite can happen after being tested in vitro and in vivo. Therefore, further studies are needed to ensure the effectiveness of the vaccine to be developed. Although epitope-based peptide vaccines have the disadvantage of low immunogenicity, the addition of adjuvants can be a solution.
Collapse
|
23
|
Lei L, Huang D, Gao H, He B, Cao J, Peppas NA. Hydrogel-guided strategies to stimulate an effective immune response for vaccine-based cancer immunotherapy. SCIENCE ADVANCES 2022; 8:eadc8738. [PMID: 36427310 PMCID: PMC9699680 DOI: 10.1126/sciadv.adc8738] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/07/2022] [Indexed: 05/25/2023]
Abstract
Cancer vaccines have attracted widespread interest in tumor therapy because of the potential to induce an effective antitumor immune response. However, many challenges including weak immunogenicity, off-target effects, and immunosuppressive microenvironments have prevented their broad clinical translation. To overcome these difficulties, effective delivery systems have been designed for cancer vaccines. As carriers in cancer vaccine delivery systems, hydrogels have gained substantial attention because they can encapsulate a variety of antigens/immunomodulators and protect them from degradation. This enables hydrogels to simultaneously reverse immunosuppression and stimulate the immune response. Meanwhile, the controlled release properties of hydrogels allow for precise temporal and spatial release of loads in situ to further enhance the immune response of cancer vaccines. Therefore, this review summarizes the classification of cancer vaccines, highlights the strategies of hydrogel-based cancer vaccines, and provides some insights into the future development of hydrogel-based cancer vaccines.
Collapse
Affiliation(s)
- Lei Lei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Dennis Huang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX 78712, USA
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Jun Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Nicholas A. Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
- Departments of Pediatrics, Surgery, and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
24
|
Huang S, Zhu Y, Zhang L, Zhang Z. Recent Advances in Delivery Systems for Genetic and Other Novel Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107946. [PMID: 34914144 DOI: 10.1002/adma.202107946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/11/2021] [Indexed: 06/14/2023]
Abstract
Vaccination is one of the most successful and cost-effective prophylactic measures against diseases, especially infectious diseases including smallpox and polio. However, the development of effective prophylactic or therapeutic vaccines for other diseases such as cancer remains challenging. This is often due to the imprecise control of vaccine activity in vivo which leads to insufficient/inappropriate immune responses or short immune memory. The development of new vaccine types in recent decades has created the potential for improving the protective potency against these diseases. Genetic and subunit vaccines are two major categories of these emerging vaccines. Owing to their nature, they rely heavily on delivery systems with various functions, such as effective cargo protection, immunogenicity enhancement, targeted delivery, sustained release of antigens, selective activation of humoral and/or cellular immune responses against specific antigens, and reduced adverse effects. Therefore, vaccine delivery systems may significantly affect the final outcome of genetic and other novel vaccines and are vital for their development. This review introduces these studies based on their research emphasis on functional design or administration route optimization, presents recent progress, and discusses features of new vaccine delivery systems, providing an overview of this field.
Collapse
Affiliation(s)
- Shiqi Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Yining Zhu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
25
|
Exosome transportation-mediated immunosuppression relief through cascade amplification for enhanced apoptotic body vaccination. Acta Biomater 2022; 153:529-539. [PMID: 36113726 DOI: 10.1016/j.actbio.2022.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/09/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022]
Abstract
Cancer vaccines represent the most promising strategies in the battle against cancers. Eliciting a robust therapeutic effect with vaccines, however, remains a challenge owing to the weak immunogenicity of autologous tumor antigens and highly immunosuppressive microenvironment. In the present study, we constructed CpG oligodeoxyribonucleotide (CpG ODN)-loaded cancer cell apoptotic bodies (Abs) as cancer vaccines for enhanced immunotherapy through cascade amplification-mediated immunosuppression relief. Abs that contain an abundant source of tumor-specific neoantigens and other tumor-associated antigens (TAAs) can be regarded as vaccines with higher immunogenicity. The de novo synthesized Abs-CpG could target and polarize macrophages to improve the immunosuppressive microenvironment. More importantly, we found that the effect of immunosuppression relief was cascade amplified, which was mediated by M1 macrophage-derived exosome transportation. Our results showed that CpG ODN polarized macrophages to M1 type and produced a large amount of TNF-α, which then activated cell division control protein 42 (Cdc42). Interestingly, we found that exosomes from M1 macrophages delivered Cdc42 and CpG to adjacent macrophages and further enhanced the phagocytosis of adjacent macrophages by positive feedback. Through cascade amplification induced by Abs-CpG with macrophage exosomes, the immunogenicity and immunosuppressive microenvironment were greatly improved, which then enhanced the performance of cancer vaccine therapy. Thus, we propose that a strategy of combining the Abs-based vaccine platform with the immunomodulatory approach represents the next generation of cancer immunotherapy. STATEMENT OF SIGNIFICANCE: 1. We discovered a relieving strategy for tumor immunosuppressive microenvironment: Abs-CpG polarized macrophages to M1 type, and M1 macrophage-derived exosomes delivered Cdc42 and CpG to adjacent macrophages, which then further enhanced the phagocytosis of adjacent macrophages by positive feedback. Through cascade amplification induced by the transfer of macrophage exosomes, the immunogenicity and immunosuppressive microenvironment were greatly improved. 2. As a vaccine, Abs contained both tumor-specific neoantigens and other tumor-associated antigens with higher immunogenicity and high clinical transformability.
Collapse
|
26
|
Giotta Lucifero A, Luzzi S. Emerging immune-based technologies for high-grade gliomas. Expert Rev Anticancer Ther 2022; 22:957-980. [PMID: 35924820 DOI: 10.1080/14737140.2022.2110072] [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: 11/04/2022]
Abstract
INTRODUCTION The selection of a tailored and successful strategy for high-grade gliomas (HGGs) treatment is still a concern. The abundance of aberrant mutations within the heterogenic genetic landscape of glioblastoma strongly influences cell expansion, proliferation, and therapeutic resistance. Identification of immune evasion pathways opens the way to novel immune-based strategies. This review intends to explore the emerging immunotherapies for HGGs. The immunosuppressive mechanisms related to the tumor microenvironment and future perspectives to overcome glioma immunity barriers are also debated. AREAS COVERED An extensive literature review was performed on the PubMed/Medline and ClinicalTrials.gov databases. Only highly relevant articles in English and published in the last 20 years were selected. Data about immunotherapies coming from preclinical and clinical trials were summarized. EXPERT OPINION The overall level of evidence about the efficacy and safety of immunotherapies for HGGs is noteworthy. Monoclonal antibodies have been approved as second-line treatment, while peptide vaccines, viral gene strategies, and adoptive technologies proved to boost a vivid antitumor immunization. Malignant brain tumor-treating fields are ever-changing in the upcoming years. Constant refinements and development of new routes of drug administration will permit to design of novel immune-based treatment algorithms thus improving the overall survival.
Collapse
Affiliation(s)
- Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.,Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
27
|
Davodabadi F, Sarhadi M, Arabpour J, Sargazi S, Rahdar A, Díez-Pascual AM. Breast cancer vaccines: New insights into immunomodulatory and nano-therapeutic approaches. J Control Release 2022; 349:844-875. [PMID: 35908621 DOI: 10.1016/j.jconrel.2022.07.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
Breast cancer (BC) is known to be a highly heterogeneous disease that is clinically subdivided into four primary molecular subtypes, each having distinct morphology and clinical implications. These subtypes are principally defined by hormone receptors and other proteins involved (or not involved) in BC development. BC therapeutic vaccines [including peptide-based vaccines, protein-based vaccines, nucleic acid-based vaccines (DNA/RNA vaccines), bacterial/viral-based vaccines, and different immune cell-based vaccines] have emerged as an appealing class of cancer immunotherapeutics when used alone or combined with other immunotherapies. Employing the immune system to eliminate BC cells is a novel therapeutic modality. The benefit of active immunotherapies is that they develop protection against neoplastic tissue and readjust the immune system to an anti-tumor monitoring state. Such immunovaccines have not yet shown effectiveness for BC treatment in clinical trials. In recent years, nanomedicines have opened new windows to increase the effectiveness of vaccinations to treat BC. In this context, some nanoplatforms have been designed to efficiently deliver molecular, cellular, or subcellular vaccines to BC cells, increasing the efficacy and persistence of anti-tumor immunity while minimizing undesirable side effects. Immunostimulatory nano-adjuvants, liposomal-based vaccines, polymeric vaccines, virus-like particles, lipid/calcium/phosphate nanoparticles, chitosan-derived nanostructures, porous silicon microparticles, and selenium nanoparticles are among the newly designed nanostructures that have been used to facilitate antigen internalization and presentation by antigen-presenting cells, increase antigen stability, enhance vaccine antigenicity and remedial effectivity, promote antigen escape from the endosome, improve cytotoxic T lymphocyte responses, and produce humoral immune responses in BC cells. Here, we summarized the existing subtypes of BC and shed light on immunomodulatory and nano-therapeutic strategies for BC vaccination. Finally, we reviewed ongoing clinical trials on BC vaccination and highlighted near-term opportunities for moving forward.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran.
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran.
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
28
|
Tang M, Cai JH, Diao HY, Guo WM, Yang X, Xing S. The progress of peptide vaccine clinical trials in gynecologic oncology. Hum Vaccin Immunother 2022; 18:2062982. [PMID: 35687860 PMCID: PMC9450897 DOI: 10.1080/21645515.2022.2062982] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Peptide vaccine are a type of immunotherapy that are synthesized according to the amino acid sequence of known or predicted tumor antigen epitopes. They are safe and well tolerated and have shown exciting results in gynecologic oncology. However, no peptide vaccine has yet been licensed in this field. This review examines peptide vaccine clinical trials in gynecology registered on ClinicalTrials.gov through January 1, 2022, analyzes the global progress and current achievements of peptide vaccines in gynecology, and explores the efforts focused on devising new methods to boost immunotherapeutic outcomes, including the use of adjuvants, multi-epitope vaccines, combinations of helper T cell epitopes, personalized peptide vaccines, synthetic long peptides, new peptide delivery, and combination therapy.
Collapse
Affiliation(s)
- Mi Tang
- GCP institution, Chengdu Women's and Children's Center Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Jiang-Hui Cai
- Department of Pharmacy, Chengdu Women's and Children's Center Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Hao-Yang Diao
- GCP institution, Chengdu Women's and Children's Center Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Wen-Mei Guo
- GCP institution, Chengdu Women's and Children's Center Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Xiao Yang
- Obstetrics Department, Chengdu Women's and Children's Center Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - ShaSha Xing
- GCP institution, Chengdu Women's and Children's Center Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| |
Collapse
|
29
|
Fang FY, Rosenblum JS, Ho WS, Heiss JD. New Developments in the Pathogenesis, Therapeutic Targeting, and Treatment of Pediatric Medulloblastoma. Cancers (Basel) 2022; 14:cancers14092285. [PMID: 35565414 PMCID: PMC9100249 DOI: 10.3390/cancers14092285] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/26/2022] [Accepted: 05/01/2022] [Indexed: 01/25/2023] Open
Abstract
Pediatric medulloblastoma (MB) is the most common pediatric brain tumor with varying prognoses depending on the distinct molecular subtype. The four consensus subgroups are WNT, Sonic hedgehog (SHH), Group 3, and Group 4, which underpin the current 2021 WHO classification of MB. While the field of knowledge for treating this disease has significantly advanced over the past decade, a deeper understanding is still required to improve the clinical outcomes for pediatric patients, who are often vulnerable in ways that adult patients are not. Here, we discuss how recent insights into the pathogenesis of pediatric medulloblastoma have directed current and future research. This review highlights new developments in understanding the four molecular subtypes’ pathophysiology, epigenetics, and therapeutic targeting. In addition, we provide a focused discussion of recent developments in imaging, and in the surgery, chemotherapy, and radiotherapy of pediatric medulloblastoma. The article includes a brief explanation of healthcare costs associated with medulloblastoma treatment.
Collapse
Affiliation(s)
- Francia Y. Fang
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Jared S. Rosenblum
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Winson S. Ho
- Department of Neurosurgery, The University of Texas at Austin, Austin, TX 78712, USA;
| | - John D. Heiss
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Correspondence:
| |
Collapse
|
30
|
Development of Peptide-Based Vaccines for Cancer. JOURNAL OF ONCOLOGY 2022; 2022:9749363. [PMID: 35342400 PMCID: PMC8941562 DOI: 10.1155/2022/9749363] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Peptides cancer vaccines are designed based on the epitope peptides that can elicit humoral and cellular immune responses targeting tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs). In order to develop a clinically safe and more effective vaccine for the future, several issues need to be addressed, and these include the selection of optimal antigen targets, adjuvants, and immunization regimens. Another emerging approach involves the use of personalized peptide-based vaccines based on neoantigens to enhance antitumor response. Rationally designed combinatorial therapy is currently being investigated with chemotherapeutic drugs or immune checkpoint inhibitor therapies to improve the efficacy. This review discusses an overview of the development of peptide-based vaccines, the role of adjuvants, and the delivery systems for peptide vaccines as well as combinatorial therapy as potential anticancer strategies.
Collapse
|
31
|
Roesler AS, Anderson KS. Beyond Sequencing: Prioritizing and Delivering Neoantigens for Cancer Vaccines. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2410:649-670. [PMID: 34914074 DOI: 10.1007/978-1-0716-1884-4_35] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neoantigens are tumor-specific proteins and peptides that can be highly immunogenic. Immune-mediated tumor rejection is strongly associated with cytotoxic responses to neoantigen-derived peptides in noncovalent association with self-HLA molecules. Neoantigen-based therapies, such as adoptive T cell transfer, have shown the potential to induce remission of treatment-resistant metastatic disease in select patients. Cancer vaccines are similarly designed to elicit or amplify antigen-specific T cell populations and stimulate directed antitumor immunity, but the selection and prioritization of the neoantigens remains a challenge. Bioinformatic algorithms can predict tumor neoantigens from somatic mutations, insertion-deletions, and other aberrant peptide products, but this often leads to hundreds of potential neoepitopes, all unique for that tumor. Selecting neoantigens for cancer vaccines is complicated by the technical challenges of neoepitope discovery, the diversity of HLA molecules, and intratumoral heterogeneity of passenger mutations leading to immune escape. Despite strong preclinical evidence, few neoantigen cancer vaccines tested in vivo have generated epitope-specific T cell populations, suggesting suboptimal immune system activation. In this chapter, we review factors affecting the prioritization and delivery of candidate neoantigens in the design of therapeutic and preventive cancer vaccines and consider synergism with standard chemotherapies.
Collapse
Affiliation(s)
- Alexander S Roesler
- School of Medicine, Duke University, Durham, NC, USA
- Mayo Clinic, Scottsdale, AZ, USA
| | - Karen S Anderson
- Mayo Clinic, Scottsdale, AZ, USA.
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
32
|
Mohsen MO, Speiser DE, Michaux J, Pak H, Stevenson BJ, Vogel M, Inchakalody VP, de Brot S, Dermime S, Coukos G, Bassani-Sternberg M, Bachmann MF. Bedside formulation of a personalized multi-neoantigen vaccine against mammary carcinoma. J Immunother Cancer 2022; 10:jitc-2021-002927. [PMID: 35017147 PMCID: PMC8753436 DOI: 10.1136/jitc-2021-002927] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 12/28/2022] Open
Abstract
Background Harnessing the immune system to purposely recognize and destroy tumors represents a significant breakthrough in clinical oncology. Non-synonymous mutations (neoantigenic peptides) were identified as powerful cancer targets. This knowledge can be exploited for further improvements of active immunotherapies, including cancer vaccines, as T cells specific for neoantigens are not attenuated by immune tolerance mechanism and do not harm healthy tissues. The current study aimed at developing an optimized multitarget vaccine using short or long neoantigenic peptides utilizing virus-like particles (VLPs) as an efficient vaccine platform. Methods Mutations of murine mammary carcinoma cells were identified by integrating mass spectrometry-based immunopeptidomics and whole exome sequencing. Neoantigenic peptides were synthesized and covalently linked to virus-like nanoparticles using a Cu-free click chemistry method for easy preparation of vaccines against mouse mammary carcinoma. Results As compared with short peptides, vaccination with long peptides was superior in the generation of neoantigen-specific CD4+ and CD8+ T cells, which readily produced interferon gamma (IFN-γ) and tumor-necrosis factor α (TNF-α). The resulting anti-tumor effect was associated with favorable immune re-polarization in the tumor microenvironment through reduction of myeloid-derived suppressor cells. Vaccination with long neoantigenic peptides also decreased post-surgical tumor recurrence and metastases, and prolonged mouse survival, despite the tumor’s low mutational burden. Conclusion Integrating mass spectrometry-based immunopeptidomics and whole exome sequencing is an efficient approach for identifying neoantigenic peptides. Our multitarget VLP-based vaccine shows a promising anti-tumor effect in an aggressive murine mammary carcinoma model. Future clinical application using this strategy is readily feasible and practical, as click chemistry coupling of personalized synthetic peptides to the nanoparticles can be done at the bedside directly before injection.
Collapse
Affiliation(s)
- Mona O Mohsen
- Department of Medical Oncology, Hamad Medical Corporation, Doha, Qatar .,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Daniel E Speiser
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Justine Michaux
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - HuiSong Pak
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | | | - Monique Vogel
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | | | - Said Dermime
- Department of Medical Oncology, National Center for Cancer Care and Research, Doha, Qatar
| | - Georges Coukos
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland .,University of Lausanne, Lausanne, Switzerland
| | - Martin F Bachmann
- Department of BioMedical Research, University of Bern, Bern, Switzerland.,Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Lu X, Bao L, Pan Z, Ge M. Immunotherapy for anaplastic thyroid carcinoma: the present and future. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:675-684. [PMID: 35347912 PMCID: PMC8931605 DOI: 10.3724/zdxbyxb-2021-0273] [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] [Received: 08/10/2021] [Accepted: 10/20/2021] [Indexed: 05/25/2023]
Abstract
Anaplastic thyroid carcinoma (ATC) is the most malignant tumor of endocrine system, which is an urgent medical problem to be solved. At present, immunotherapy studies on ATC mainly include cutting off the recruitment of tumor-associated macrophage (TAM), inducing the reprogramming of TAM and restoring its phagocytic function, targeting related immune checkpoints on T cells and natural killer cells, tumor vaccines based on oncolytic viruses and dendritic cells, and adoptive immunotherapy. Among them, immunotherapy strategies represented by targeted blocking of programmed death-1/programmed death ligand-1 at immune checkpoint have been preliminarily confirmed to benefit ATC patients, especially the combination of molecular targeted inhibitors and immunotherapy has shown excellent therapeutic effects. Due to the great heterogeneity of ATC, it is expected to provide more therapeutic strategies for patients of ATC by carrying out various immunotherapy studies including biological, immune and cellular therapies and exploring the therapeutic potential of the next generation of immune checkpoint inhibitors. This article reviews the potential immunotherapeutic targets of ATC and the progress of immunotherapy.
Collapse
Affiliation(s)
- Xixuan Lu
- 1. Department of Head and Neck Surgery, Center of Otolaryngology, Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
- 2. Zhejiang Provincial Key Laboratory of Endocrine Gland Diseases, Hangzhou 310014, China
| | - Lisha Bao
- 1. Department of Head and Neck Surgery, Center of Otolaryngology, Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
- 2. Zhejiang Provincial Key Laboratory of Endocrine Gland Diseases, Hangzhou 310014, China
| | - Zongfu Pan
- 2. Zhejiang Provincial Key Laboratory of Endocrine Gland Diseases, Hangzhou 310014, China
- 3. Department of Pharmacy, Clinical Pharmacy Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Minghua Ge
- 1. Department of Head and Neck Surgery, Center of Otolaryngology, Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
- 2. Zhejiang Provincial Key Laboratory of Endocrine Gland Diseases, Hangzhou 310014, China
| |
Collapse
|
34
|
Wang EJ, Chen JS, Jain S, Morshed RA, Haddad AF, Gill S, Beniwal AS, Aghi MK. Immunotherapy Resistance in Glioblastoma. Front Genet 2021; 12:750675. [PMID: 34976006 PMCID: PMC8718605 DOI: 10.3389/fgene.2021.750675] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults. Despite treatment consisting of surgical resection followed by radiotherapy and adjuvant chemotherapy, survival remains poor at a rate of 26.5% at 2 years. Recent successes in using immunotherapies to treat a number of solid and hematologic cancers have led to a growing interest in harnessing the immune system to target glioblastoma. Several studies have examined the efficacy of various immunotherapies, including checkpoint inhibitors, vaccines, adoptive transfer of lymphocytes, and oncolytic virotherapy in both pre-clinical and clinical settings. However, these therapies have yielded mixed results at best when applied to glioblastoma. While the initial failures of immunotherapy were thought to reflect the immunoprivileged environment of the brain, more recent studies have revealed immune escape mechanisms created by the tumor itself and adaptive resistance acquired in response to therapy. Several of these resistance mechanisms hijack key signaling pathways within the immune system to create a protumoral microenvironment. In this review, we discuss immunotherapies that have been trialed in glioblastoma, mechanisms of tumor resistance, and strategies to sensitize these tumors to immunotherapies. Insights gained from the studies summarized here may help pave the way for novel therapies to overcome barriers that have thus far limited the success of immunotherapy in glioblastoma.
Collapse
Affiliation(s)
- Elaina J. Wang
- Department of Neurological Surgery, The Warren Alpert School of Medicine, Brown University, Providence, RI, United States
| | - Jia-Shu Chen
- Department of Neurological Surgery, The Warren Alpert School of Medicine, Brown University, Providence, RI, United States
| | - Saket Jain
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Ramin A. Morshed
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander F. Haddad
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Sabraj Gill
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Angad S. Beniwal
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Manish K. Aghi
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
35
|
Sun Z, Sun X, Chen Z, Du J, Wu Y. Head and Neck Squamous Cell Carcinoma: Risk Factors, Molecular Alterations, Immunology and Peptide Vaccines. Int J Pept Res Ther 2021; 28:19. [PMID: 34903958 PMCID: PMC8653808 DOI: 10.1007/s10989-021-10334-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 12/29/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) arises from the epithelial lining of the oral cavity, hypopharynx, oropharynx, and larynx. There are several potential risk factors that cause the generation of HNSCC, including cigarette smoking, alcohol consumption, betel quid chewing, inadequate nutrition, poor oral hygiene, HPV and Epstein–Barr virus, and Candida albicans infections. HNSCC has causative links to both environmental factors and genetic mutations, with the latter playing a more critical role in cancer progression. These molecular changes to epithelial cells include the inactivation of cancer suppressor genes and proto-oncogenes overexpression, resulting in tumour cell proliferation and distant metastasis. HNSCC patients have impaired dendritic cell (DC) and natural killer (NK) cell functions, increased production of higher immune-suppressive molecules, loss of regulatory T cells and co-stimulatory molecules and major histocompatibility complex (MHC) class Ι molecules, lower number of lymphocyte subsets, and a poor response to antigen-presenting cells. At present, the standard treatment modalities for HNSCC patients include surgery, chemotherapy and radiotherapy, and combinatorial therapy. Despite advances in the development of novel treatment modalities over the last few decades, survival rates of HNSCC patients have not increased. To establish effective immunotherapies, a greater understanding of interactions between the immune system and HNSCC is required, and there is a particular need to develop novel therapeutic options. A therapeutic cancer vaccine has been proposed as a promising method to improve outcome by inducing a powerful adaptive immune response that leads to cancer cell elimination. Compared with other vaccines, peptide cancer vaccines are more robust and specific. In the past few years, there have been remarkable achievements in peptide-based vaccines for HNSCC patients. Here, we summarize the latest molecular alterations in HNSCC, explore the immune response to HNSCC, and discuss the latest developments in peptide-based cancer vaccine strategies. This review highlights areas for valuable future research focusing on peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, Shandong 250000 China
| | - Zhanwei Chen
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| | - Juan Du
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| | - Yihua Wu
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| |
Collapse
|
36
|
Munhoz J, Thomé R, Rostami A, Ishikawa LLW, Verinaud L, Rapôso C. The SNX-482 peptide from Hysterocrates gigas spider acts as an immunomodulatory molecule activating macrophages. Peptides 2021; 146:170648. [PMID: 34537257 DOI: 10.1016/j.peptides.2021.170648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
Peptides are molecules that have emerged as crucial candidates for the development of anticancer drugs. Spider venoms are a rich source of peptides (venom peptides - VPs) with biological effects. VPs have been tested as adjuvants in the activation of cells of the immune system with the aim of improving immunotherapies for the treatment of neoplasms. In the present study, the effects of SNX-482, a peptide from the African tarantula Hysterocrates gigas, on macrophages were described. The results showed that the peptide activated M0-macrophages, increasing costimulatory molecules (CD40, CD68, CD80, CD83, CD86) involved in antigen presentation, and also augmenting the checkpoint molecules PD-L1, CTLA-4 and FAS-L; these effects were not concentration-dependent. SNX-482 also increased the release of IL-23 and upregulated the expression of ccr4, ifn-g, gzmb and pdcd1, genes important for the anticancer response. The pretreatment of macrophages with the peptide did not interfere in the modulation of T cells, and macrophages previously polarized to M1 and M2 profile did not respond to SNX-482. These findings represent the expansion of knowledge about the use of VPs in drug discovery, pointing to a potential new candidate for anticancer immunotherapy. Considering that most immunotherapies target the adaptive system, the modulation of macrophages (an innate immune cell) by SNX-482 is especially relevant.
Collapse
Affiliation(s)
- Jaqueline Munhoz
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Rodolfo Thomé
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | - Liana Verinaud
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, Brazil
| | - Catarina Rapôso
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil; Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, Brazil.
| |
Collapse
|
37
|
Antonarelli G, Corti C, Tarantino P, Ascione L, Cortes J, Romero P, Mittendorf EA, Disis ML, Curigliano G. Therapeutic cancer vaccines revamping: technology advancements and pitfalls. Ann Oncol 2021; 32:1537-1551. [PMID: 34500046 PMCID: PMC8420263 DOI: 10.1016/j.annonc.2021.08.2153] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/21/2021] [Accepted: 08/29/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer vaccines (CVs) represent a long-sought therapeutic and prophylactic immunotherapy strategy to obtain antigen (Ag)-specific T-cell responses and potentially achieve long-term clinical benefit. However, historically, most CV clinical trials have resulted in disappointing outcomes, despite promising signs of immunogenicity across most formulations. In the past decade, technological advances regarding vaccine delivery platforms, tools for immunogenomic profiling, and Ag/epitope selection have occurred. Consequently, the ability of CVs to induce tumor-specific and, in some cases, remarkable clinical responses have been observed in early-phase clinical trials. It is notable that the record-breaking speed of vaccine development in response to the coronavirus disease-2019 pandemic mainly relied on manufacturing infrastructures and technological platforms already developed for CVs. In turn, research, clinical data, and infrastructures put in place for the severe acute respiratory syndrome coronavirus 2 pandemic can further speed CV development processes. This review outlines the main technological advancements as well as major issues to tackle in the development of CVs. Possible applications for unmet clinical needs will be described, putting into perspective the future of cancer vaccinology.
Collapse
Affiliation(s)
- G Antonarelli
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - C Corti
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - P Tarantino
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - L Ascione
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - J Cortes
- International Breast Cancer Center (IBCC), Quironsalud Group, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - P Romero
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
| | - E A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, USA; Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, USA
| | - M L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, USA
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
38
|
Lemon JL, McMenamy MJ. A Review of UK-Registered and Candidate Vaccines for Bovine Respiratory Disease. Vaccines (Basel) 2021; 9:vaccines9121403. [PMID: 34960149 PMCID: PMC8703677 DOI: 10.3390/vaccines9121403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/10/2021] [Accepted: 11/22/2021] [Indexed: 01/11/2023] Open
Abstract
Vaccination is widely regarded as a cornerstone in animal or herd health and infectious disease management. Nineteen vaccines against the major pathogens implicated in bovine respiratory disease are registered for use in the UK by the Veterinary Medicines Directorate (VMD). However, despite annual prophylactic vaccination, bovine respiratory disease is still conservatively estimated to cost the UK economy approximately £80 million per annum. This review examines the vaccine types available, discusses the surrounding literature and scientific rationale of the limitations and assesses the potential of novel vaccine technologies.
Collapse
Affiliation(s)
- Joanne L. Lemon
- Sustainable Agri-Food and Sciences Division, Agri-Food and Bioscience Institute, Newforge Lane, Belfast BT9 5PX, UK
- Correspondence:
| | - Michael J. McMenamy
- Veterinary Sciences Division, Agri-Food and Bioscience Institute, Stormont, Belfast BT4 3SD, UK;
| |
Collapse
|
39
|
Biological Therapies in the Treatment of Cancer-Update and New Directions. Int J Mol Sci 2021; 22:ijms222111694. [PMID: 34769123 PMCID: PMC8583892 DOI: 10.3390/ijms222111694] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Biological therapies have changed the face of oncology by targeting cancerous cells while reducing the effect on normal tissue. This publication focuses mainly on new therapies that have contributed to the advances in treatment of certain malignancies. Immunotherapy, which has repeatedly proven to be a breakthrough therapy in melanoma, as well as B-ALL therapy with CAR T cells, are of great merit in this progress. These therapies are currently being developed by modifying bispecific antibodies and CAR T cells to improve their efficiency and bioavailability. Work on improving the therapy with oncolytic viruses is also progressing, and efforts are being made to improve the immunogenicity and stability of cancer vaccines. Combining various biological therapies, immunotherapy with oncolytic viruses or cancer vaccines is gaining importance in cancer therapy. New therapeutic targets are intensively sought among neoantigens, which are not immunocompromised, or antigens associated with tumor stroma cells. An example is fibroblast activation protein α (FAPα), the overexpression of which is observed in the case of tumor progression. Universal therapeutic targets are also sought, such as the neurotrophic receptor tyrosine kinase (NTRK) gene fusion, a key genetic driver present in many types of cancer. This review also raises the problem of the tumor microenvironment. Stromal cells can protect tumor cells from chemotherapy and contribute to relapse and progression. This publication also addresses the problem of cancer stem cells resistance to treatment and presents attempts to avoid this phenomenon. This review focuses on the most important strategies used to improve the selectivity of biological therapies.
Collapse
|
40
|
Ikezaki M, Nishitsuji K, Matsumura K, Manabe S, Shibukawa Y, Wada Y, Ito Y, Ihara Y. C-Mannosylated tryptophan-containing WSPW peptide binds to actinin-4 and alters E-cadherin subcellular localization in lung epithelial-like A549 cells. Biochimie 2021; 192:136-146. [PMID: 34673139 DOI: 10.1016/j.biochi.2021.10.007] [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: 04/12/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022]
Abstract
The Trp-x-x-Trp (W-x-x-W) peptide motif, a consensus site for C-mannosylation, is the functional motif in cytokine type I receptors or thrombospondin type I repeat (TSR) superfamily proteins. W-x-x-W motifs are important for physiological and pathological functions of their parental proteins, but effects of C-mannosylation on protein functions remain to be elucidated. By using chemically synthesized WSPW peptides and C-mannosylated WSPW peptides (C-Man-WSPW), we herein investigated whether C-mannosylation of WSPW peptides confer additional biological functions to WSPW peptides. C-Man-WSPW peptide, but not non-mannosylated WSPW, reduced E-cadherin levels in A549 cells. Via peptide mass fingerprinting analysis, we identified actinin-4 as a C-Man-WSPW-binding protein in A549 cells. Actinin-4 partly co-localized with E-cadherin or β-catenin, despite no direct interaction between actinin-4 and E-cadherin. C-Man-WSPW reduced co-localization of E-cadherin and actinin-4; non-mannosylated WSPW had no effect on localization. In actinin-4-knockdown cells, E-cadherin was upregulated and demonstrated a punctate staining pattern in the cytoplasm, which suggests that actinin-4 regulated cell-surface E-cadherin localization. Thus, C-mannosylation of WSPW peptides is required for interaction with actinin-4 that subsequently alters expression and subcellular localization of E-cadherin and morphology of epithelial-like cells. Our results therefore suggest a regulatory role of C-mannosylation of the W-x-x-W motif in interactions between the motif and its binding partner and will thereby enhance understanding of protein C-mannosylation.
Collapse
Affiliation(s)
- Midori Ikezaki
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan
| | - Kazuchika Nishitsuji
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan.
| | - Ko Matsumura
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan
| | - Shino Manabe
- Laboratory of Functional Molecule Chemistry, Pharmaceutical Department and Institute of Medicinal Chemistry, Hoshi University, Tokyo, 142-8501, Japan; Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Tohoku University, Miyagi, 980-8578, Japan
| | - Yukinao Shibukawa
- Department of Molecular Medicine, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, 594-1101, Japan
| | - Yoshinao Wada
- Department of Molecular Medicine, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, 594-1101, Japan
| | - Yukishige Ito
- RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan; Graduate School of Science, Osaka University, Osaka, 560-0043, Japan
| | - Yoshito Ihara
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan.
| |
Collapse
|
41
|
Wilks LR, Joshi G, Grisham MR, Gill HS. Tyrosine-Based Cross-Linking of Peptide Antigens to Generate Nanoclusters with Enhanced Immunogenicity: Demonstration Using the Conserved M2e Peptide of Influenza A. ACS Infect Dis 2021; 7:2723-2735. [PMID: 34432416 DOI: 10.1021/acsinfecdis.1c00219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A method of creating nanoclusters (NCs) from soluble peptide molecules is described utilizing an approach based on a tyrosine-tyrosine cross-linking reaction. A reactive tag comprising histidine and tyrosine residues was introduced at the termini of the peptide molecules. The cross-linking reaction led to the creation of dityrosine bonds within the tag, which allowed for the generation of peptide NCs. We show that it is essential for the reactive tag to be present at both the "N" and "C" termini of the peptide for cluster formation to occur. Additionally, the cross-linking reaction was systematically characterized to show the importance of reaction conditions on final cluster diameter, allowing us to generate NCs of various sizes. To demonstrate the immunogenic potential of the peptide clusters, we chose to study the conserved influenza peptide, M2e, as the antigen. M2e NCs were formulated using the cross-linking reaction. We show the ability of the clusters to generate protective immunity in a dose, size, and frequency dependent manner against a lethal influenza A challenge in BALB/c mice. Taken together, the data presented suggest this new cluster formation technique can generate highly immunogenic peptide NCs in a simple and controllable manner.
Collapse
Affiliation(s)
- Logan R. Wilks
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Megan R. Grisham
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
42
|
Lee D, Huntoon K, Wang Y, Jiang W, Kim BYS. Harnessing Innate Immunity Using Biomaterials for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007576. [PMID: 34050699 DOI: 10.1002/adma.202007576] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/29/2021] [Indexed: 06/12/2023]
Abstract
The discovery of immune checkpoint blockade has revolutionized the field of immuno-oncology and established the foundation for developing various new therapies that can surpass conventional cancer treatments. Most recent immunotherapeutic strategies have focused on adaptive immune responses by targeting T cell-activating pathways, genetic engineering of T cells with chimeric antigen receptors, or bispecific antibodies. Despite the unprecedented clinical success, these T cell-based treatments have only benefited a small proportion of patients. Thus, the need for the next generation of cancer immunotherapy is driven by identifying novel therapeutic molecules or new immunoengineered cells. To maximize the therapeutic potency via innate immunogenicity, the convergence of innate immunity-based therapy and biomaterials is required to yield an efficient index in clinical trials. This review highlights how biomaterials can efficiently reprogram and recruit innate immune cells in tumors and ultimately initiate activation of T cell immunity against advanced cancers. Moreover, the design and specific biomaterials that improve innate immune cells' targeting ability to selectively activate immunogenicity with minimal adverse effects are discussed.
Collapse
Affiliation(s)
- DaeYong Lee
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
43
|
Cancer Vaccines: Promising Therapeutics or an Unattainable Dream. Vaccines (Basel) 2021; 9:vaccines9060668. [PMID: 34207062 PMCID: PMC8233841 DOI: 10.3390/vaccines9060668] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 02/08/2023] Open
Abstract
The advent of cancer immunotherapy has revolutionized the field of cancer treatment and offers cancer patients new hope. Although this therapy has proved highly successful for some patients, its efficacy is not all encompassing and several cancer types do not respond. Cancer vaccines offer an alternate approach to promote anti-tumor immunity that differ in their mode of action from antibody-based therapies. Cancer vaccines serve to balance the equilibrium of the crosstalk between the tumor cells and the host immune system. Recent advances in understanding the nature of tumor-mediated tolerogenicity and antigen presentation has aided in the identification of tumor antigens that have the potential to enhance anti-tumor immunity. Cancer vaccines can either be prophylactic (preventative) or therapeutic (curative). An exciting option for therapeutic vaccines is the emergence of personalized vaccines, which are tailor-made and specific for tumor type and individual patient. This review summarizes the current standing of the most promising vaccine strategies with respect to their development and clinical efficacy. We also discuss prospects for future development of stem cell-based prophylactic vaccines.
Collapse
|
44
|
Rahimian N, Miraei HR, Amiri A, Ebrahimi MS, Nahand JS, Tarrahimofrad H, Hamblin MR, Khan H, Mirzaei H. Plant-based vaccines and cancer therapy: Where are we now and where are we going? Pharmacol Res 2021; 169:105655. [PMID: 34004270 DOI: 10.1016/j.phrs.2021.105655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
Therapeutic vaccines are an effective approach in cancer therapy for treating the disease at later stages. The Food and Drug Administration (FDA) recently approved the first therapeutic cancer vaccine, and further studies are ongoing in clinical trials. These are expected to result in the future development of vaccines with relatively improved efficacy. Several vaccination approaches are being studied in pre-clinical and clinical trials, including the generation of anti-cancer vaccines by plant expression systems.This approach has advantages, such as high safety and low costs, especially for the synthesis of recombinant proteins. Nevertheless, the development of anti-cancer vaccines in plants is faced with some technical obstacles.Herein, we summarize some vaccines that have been used in cancer therapy, with an emphasis on plant-based vaccines.
Collapse
Affiliation(s)
- Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Hamid Reza Miraei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashahd, Iran
| | | | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Tarrahimofrad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 20282028, South Africa
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
45
|
Ciobanasu C. Peptides-based therapy and diagnosis. Strategies for non-invasive therapies in cancer. J Drug Target 2021; 29:1063-1079. [PMID: 33775187 DOI: 10.1080/1061186x.2021.1906885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, remarkable progress was registered in the field of cancer research. Though, cancer still represents a major cause of death and cancer metastasis a problem seeking for urgent solutions as it is the main reason for therapeutic failure. Unfortunately, the most common chemotherapeutic agents are non-selective and can damage healthy tissues and cause side effects that affect dramatically the quality of life of the patients. Targeted therapy with molecules that act specifically at the tumour sites interacting with overexpressed cancer receptors is a very promising strategy for achieving the specific delivery of anticancer drugs, radioisotopes or imaging agents. This review aims to give an overview on different strategies for targeting cancer cell receptors localised either at the extracellular matrix or at the cell membrane. Molecules like antibodies, aptamers and peptides targeting the cell surface are presented with advantages and disadvantages, with emphasis on peptides. The most representative peptides are described, including cell penetrating peptides, homing and anticancer peptides with particular consideration on recent discoveries.
Collapse
Affiliation(s)
- Corina Ciobanasu
- Sciences Department, Institute for Interdisciplinary Research, Alexandru I. Cuza University, Iaşi, Romania
| |
Collapse
|
46
|
Engineering a sustained release vaccine with a pathogen-mimicking manner for robust and durable immune responses. J Control Release 2021; 333:162-175. [PMID: 33794269 DOI: 10.1016/j.jconrel.2021.03.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Sustained release vaccine carriers can facilitate an increased interaction time between the antigen and immune system to strengthen immune responses, but their promotion on adaptive immune responses, especially cellular immunity, are still unfavorable. Herein, we report a sustained antigen delivery vector, which carries abundant antigens, a nucleic acid adjuvant and pathogen-associated molecular patterns to simulate a natural pathogen to reinforce immune responses. Specifically, murine colorectal cancer cells MC38 lysate and Toll-like receptor 9 agonist CpG are loaded into yeast derived β-glucan particles (GPs). After vaccination, these particles can form a vaccine depot that continuously release the antigen similar to the traditional aluminum hydroxide gel, but recruit more immune cells and induce more cytokine secretion at the injection site. Stronger antibody responses, Th1 and Th17 biased cellular immunity and immune memory are achieved compared with aluminum hydroxide gel. More importantly, treatment with these particles significantly suppress tumor growth in a therapeutic tumor model. This work shed light on the efficacy of combining sustained antigen release with pathogen-mimicking manner in vaccine design.
Collapse
|
47
|
Olsen HE, Lynn GM, Valdes PA, Cerecedo Lopez CD, Ishizuka AS, Arnaout O, Bi WL, Peruzzi PP, Chiocca EA, Friedman GK, Bernstock JD. Therapeutic cancer vaccines for pediatric malignancies: advances, challenges, and emerging technologies. Neurooncol Adv 2021; 3:vdab027. [PMID: 33860227 PMCID: PMC8034661 DOI: 10.1093/noajnl/vdab027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Though outcomes for pediatric cancer patients have significantly improved over the past several decades, too many children still experience poor outcomes and survivors suffer lifelong, debilitating late effects after conventional chemotherapy, radiation, and surgical treatment. Consequently, there has been a renewed focus on developing novel targeted therapies to improve survival outcomes. Cancer vaccines are a promising type of immunotherapy that leverage the immune system to mediate targeted, tumor-specific killing through recognition of tumor antigens, thereby minimizing off-target toxicity. As such, cancer vaccines are orthogonal to conventional cancer treatments and can therefore be used alone or in combination with other therapeutic modalities to maximize efficacy. To date, cancer vaccination has remained largely understudied in the pediatric population. In this review, we discuss the different types of tumor antigens and vaccine technologies (dendritic cells, peptides, nucleic acids, and viral vectors) evaluated in clinical trials, with a focus on those used in children. We conclude with perspectives on how advances in combination therapies, tumor antigen (eg, neoantigen) selection, and vaccine platform optimization can be translated into clinical practice to improve outcomes for children with cancer.
Collapse
Affiliation(s)
- Hannah E Olsen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Pablo A Valdes
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian D Cerecedo Lopez
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Omar Arnaout
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - W Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory K Friedman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Avidea Technologies, Inc., Baltimore, Maryland, USA.,Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Protective immune response against P32 oncogenic peptide-pulsed PBMCs in mouse models of breast cancer. Int Immunopharmacol 2021; 93:107414. [PMID: 33578183 DOI: 10.1016/j.intimp.2021.107414] [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] [Received: 11/25/2020] [Revised: 12/23/2020] [Accepted: 01/17/2021] [Indexed: 12/22/2022]
Abstract
High expression of p32 in certain tumors makes it a potential target for immunotherapy. In the present study, the first goal was to design multi-epitope peptides from the P32 protein and the second goal was to compare the prophylactic effects of DCs- and PBMCs- based vaccines by pulsing them with designed peptides. For these purposes, 160 BALB/c mice were vaccinated in 5 different subgroups of each 4 peptides using PBS (F1-4a), F peptides alone (F1-4b), F peptides with CpG-ODN (F1-4c), F peptides with CpGODN and DCs (F1-4d), and F peptides with CpG-ODN and PBMCs (F1-4e). We found a significantly higher interferon-γ (IFN-γ) and granzyme B levels in T cells of F4d and F4e subgroups compared to control (p ≤ 0.05). The result of challenging spleen PBMCs of vaccinated mice with 4T1 cells showed significant up- and down- regulation of Fas ligand (FasL) and forkhead box P3 (Foxp3) gene expression between F4d and F4e subgroups with control, respectively. In addition, a significant change was seen in Caspase3 gene expression of F4d subgroup compared to control (p ≤ 0.05). Supernatant levels of IFN-γ and perforin were significantly increased in F4d and F4e subgroups compared to control. Consequently, significantly lower tumor sizes and prolonged survival time were detected in F4d and F4e subgroups compared to control after challenging mice with 4T1 cells. Accordingly, these results demonstrated that PBMCs pulsed F4 peptide-based vaccine could induce a protective immune response while it is a simple and less expensive vaccine.
Collapse
|
49
|
Chen F, Wang Y, Gao J, Saeed M, Li T, Wang W, Yu H. Nanobiomaterial-based vaccination immunotherapy of cancer. Biomaterials 2021; 270:120709. [PMID: 33581608 DOI: 10.1016/j.biomaterials.2021.120709] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapies including cancer vaccines, immune checkpoint blockade or chimeric antigen receptor T cells have been exploited as the attractive treatment modalities in recent years. Among these approaches, cancer vaccines that designed to deliver tumor antigens and adjuvants to activate the antigen presenting cells (APCs) and induce antitumor immune responses, have shown significant efficacy in inhibiting tumor growth, preventing tumor relapse and metastasis. Despite the potential of cancer vaccination strategies, the therapeutic outcomes in preclinical trials are failed to promote their clinical translation, which is in part due to their inefficient vaccination cascade of five critical steps: antigen identification, antigen encapsulation, antigen delivery, antigen release and antigen presentation to T cells. In recent years, it has been demonstrated that various nanobiomaterials hold great potential to enhance cancer vaccination cascade and improve their antitumor performance and reduce the off-target effect. We summarize the cutting-edge advances of nanobiomaterials-based vaccination immunotherapy of cancer in this review. The various cancer nanovaccines including antigen peptide/adjuvant-based nanovaccines, nucleic acid-based nanovaccines as well as biomimetic nanobiomaterials-based nanovaccines are discussed in detail. We also provide some challenges and perspectives associated with the clinical translation of cancer nanovaccines.
Collapse
Affiliation(s)
- Fangmin Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingjie Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Nano Science and Technology Institute, University of Science and Technology of China, Suzhou, 215123, China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Madiha Saeed
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tianliang Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
50
|
Abstract
There are strong biologic and preclinical rationales for the development of therapeutic cancer vaccines; however, the clinical translation of this treatment strategy has been challenging. It is now understood that many previous clinical trials of cancer vaccines used target antigens or vaccine designs that inherently lacked sufficient immunogenicity to induce clinical responses. Despite the historical track record, breakthrough advances in cancer immunobiology and vaccine technologies have supported continued interest in therapeutic cancer vaccinations, with the hope that next-generation vaccine strategies will enable patients with cancer to develop long-lasting anti-tumor immunity. There has been substantial progress identifying antigens and vaccine vectors that lead to strong and broad T cell responses, tailoring vaccine designs to achieve optimal antigen presentation, and finding combination partners employing complementary mechanisms of action (e.g., checkpoint inhibitors) to overcome the diverse methods cancer cells use to evade and suppress the immune system. Results from randomized, phase 3 studies testing therapeutic cancer vaccines based on these advances are eagerly awaited. Here, we summarize the successes and failures in the clinical development of cancer vaccines, address how this historical experience and advances in science and technology have shaped efforts to improve vaccines, and offer a clinical perspective on the future role of vaccine therapies for cancer.
Collapse
|