1
|
Butkowsky C, Aldor N, Poynter SJ. Toll‑like receptor 3 ligands for breast cancer therapies (Review). Mol Clin Oncol 2023; 19:60. [PMID: 37424627 PMCID: PMC10326562 DOI: 10.3892/mco.2023.2656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/31/2023] [Indexed: 07/11/2023] Open
Abstract
Breast cancer is the most common cause of cancer worldwide and is the leading cause of mortality for women across most of the world. Immunotherapy is a burgeoning area of cancer treatment, including for breast cancer; these are therapies that harness the power of the immune system to clear cancerous cells. Toll-like receptor 3 (TLR3) is an RNA receptor found in the endosome, and ligands that bind to TLR3 are currently being tested for their efficacy as breast cancer immunotherapeutics. The current review introduces TLR3 and the role of this receptor in breast cancer, and summarizes data on the potential use of TLR3 ligands, mainly polyinosinic:polycytidylic acid and its derivatives, as breast cancer monotherapies or, more commonly, as combination therapies with chemotherapies, other immunotherapies and cancer vaccines. The current state of TLR3 ligand breast cancer therapy research is summarized by reporting on past and current clinical trials, and notable preliminary in vitro studies are discussed. In conclusion, TLR3 ligands have robust potential in anticancer applications as innate immune stimulants, and further studies combined with innovative technologies, such as nanoparticles, may contribute to their success.
Collapse
Affiliation(s)
- Carly Butkowsky
- Department of Health Sciences, Faculty of Science, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - Natalie Aldor
- Department of Health Sciences, Faculty of Science, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - Sarah J. Poynter
- Department of Health Sciences, Faculty of Science, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| |
Collapse
|
2
|
Fan L, Sui XY, Jin X, Zhang WJ, Zhou P, Shao ZM. High expression of TLR3 in triple-negative breast cancer predicts better prognosis-data from the Fudan University Shanghai Cancer Center cohort and tissue microarrays. BMC Cancer 2023; 23:298. [PMID: 37005579 PMCID: PMC10067281 DOI: 10.1186/s12885-023-10721-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/08/2023] [Indexed: 04/04/2023] Open
Abstract
INTRODUCTION We have previously reported that Toll-like receptor 3 (TLR3) acts as a suppressor gene for breast cancer initiation and progression. In this study, we evaluated the role of TLR3 in breast cancer using our original Fudan University Shanghai Cancer Center (FUSCC) datasets and breast cancer tissue microarrays. METHODS Using FUSCC multiomics datasets on triple- negative breast cancer (TNBC), we compared the mRNA expression of TLR3 in TNBC tissue and the adjacent normal tissue. A Kaplan-Meier plotter was performed to investigate the expression of TLR3 on prognosis in the FUSCC TNBC cohort. We performed immunohistochemical staining to analyze TLR3 protein expression in the TNBC tissue microarrays. Furthermore, bioinformatics analysis was performed using the Cancer Genome Atlas (TCGA) data to verify the results of our FUSCC study. The relationship between TLR3 and clinicopathological features was analyzed with logistic regression and the Wilcoxon signed-rank test. The association between clinical characteristics and overall survival in TCGA patients was assessed using the Kaplan-Meier method and Cox regression analysis. Gene set enrichment analysis (GSEA) was performed to identify signaling pathways that are differentially activated in breast cancer. RESULTS The mRNA expression of TLR3 was lower in TNBC tissue than in the adjacent normal tissue in the FUSCC datasets. The TLR3 had high expression in immunomodulatory (IM) and mesenchymal-like (MES) subtypes and low expression in luminal androgen receptor (LAR) and basal-like immune-suppressed (BLIS) subtypes. High expression of TLR3 in TNBC predicted better prognosis in the FUSCC TNBC cohort. Immunohistochemical staining of the tissue microarrays showed that TLR3 had lower expression in breast cancer tissues than in the adject normal tissues. Furthermore, the TLR3 expression was positively associated with B cell, CD4 + T cells, CD8 + T cells, neutrophils, macrophages, and myeloid dendritic cells. Bioinformatic analysis using high-throughput RNA-sequencing data from the TCGA demonstrated that the reduced expression of TLR3 in breast cancer was associated with advanced clinicopathological characteristics, survival time, and poor prognosis. CONCLUSIONS TLR3 has low expression in TNBC tissue. High expression of TLR3 in triple-negative breast cancer predicts better prognosis. TLR3 expression may be a potential prognostic molecular marker of poor survival in breast cancer.
Collapse
Affiliation(s)
- Lei Fan
- Department of Breast Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin-Yi Sui
- Department of Breast Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xi Jin
- Department of Breast Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wen-Juan Zhang
- Department of Breast Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peng Zhou
- Department of Breast Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Parkway Health, Shanghai, China.
- Runshangshan Medical Center, Shanghai, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
León-Letelier RA, Katayama H, Hanash S. Mining the Immunopeptidome for Antigenic Peptides in Cancer. Cancers (Basel) 2022; 14:4968. [PMID: 36291752 PMCID: PMC9599891 DOI: 10.3390/cancers14204968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
Although harnessing the immune system for cancer therapy has shown success, response to immunotherapy has been limited. The immunopeptidome of cancer cells presents an opportunity to discover novel antigens for immunotherapy applications. These neoantigens bind to MHC class I and class II molecules. Remarkably, the immunopeptidome encompasses protein post-translation modifications (PTMs) that may not be evident from genome or transcriptome profiling. A case in point is citrullination, which has been demonstrated to induce a strong immune response. In this review, we cover how the immunopeptidome, with a special focus on PTMs, can be utilized to identify cancer-specific antigens for immunotherapeutic applications.
Collapse
Affiliation(s)
| | | | - Sam Hanash
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
4
|
Li WH, Su JY, Li YM. Rational Design of T-Cell- and B-Cell-Based Therapeutic Cancer Vaccines. Acc Chem Res 2022; 55:2660-2671. [PMID: 36048514 DOI: 10.1021/acs.accounts.2c00360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cancer vaccines provide an efficient strategy to enhance tumor-specific immune responses by redeploying immune systems. Despite the approval of the first cancer vaccine (Sipuleucel-T) by the U.S. Food and Drug Administration in 2010, most therapeutic cancer vaccines fail in clinical trials. Basically, tumor-specific immune responses rely on not only T-cell but also B-cell immunity, which indicates that cancer vaccines should leverage both arms of the adaptive immune system. For example, CD8+ T cells activated by antigen-presenting cells (APCs) recognize and directly kill tumor cells via peptide-bound major histocompatibility complex (pMHC). B cells recognize antigen with no need of pMHC and require CD4+ T cells for sufficient activation and antibody generation, enabling antibody-mediated nondirect killing on tumor cells. Considering the different mechanisms of T-cell and B-cell activation, the rational design of therapeutic cancer vaccines should consider several factors, including antigen selection and recognition, immune activation, vaccine delivery, and repeatable vaccination, which can be advanced by chemical strategies.In this Account, we summarize our recent contributions to the development of effective T-cell- and B-cell-based therapeutic cancer vaccines. For T-cell-based vaccines, we focus on adjuvants as the key component for controllable APC activation and T-cell priming. Not only synthetic molecular agonists of pattern recognition receptors (PRRs) but also adjuvant nanomaterials were explored to satisfy diversiform vaccine designs. For example, a type of natural cyclic dinucleotide (CDN) that was chemically modified with fluorination and ipsilateral phosphorothioation to activate the stimulator of interferon gene (STING) was found to mediate antitumor responses. It retains structural similarity to the parent CDN scaffold but possesses increased stability, cellular uptake, and immune activation for antitumor treatment. It also facilitates facile conjugation with other agonists, which not only enhances APC-targeting delivery but also balances cellular and humoral antitumor responses. We also explored the intrinsic properties of nanomaterials that allow them to serve as adjuvants. A black phosphorus nanosheet-based nanovaccine was constructed and found to strongly potentiate antigen-specific T-cell antitumor immune responses through multiple immune-potentiating properties, leading to a highly integrated nanomaterial-based adjuvant design. For B-cell-based vaccines, multicomponent and multivalent strategies were applied to improve the immunogenicity. A multicomponent linear vaccine conjugate coordinates helper T (Th) cells and APCs to proliferate and differentiates B cells for enhanced antitumor immunoglobulin G antibody responses. To further improve antigen recognition, clustered designs on a multivalent epitope were applied by generating various structures, including branched lysine-based peptides, natural multivalent scaffold molecules, and self-assembled nanofibers. We also engineered nano- and microvaccine systems to optimize systemic and localized vaccination. A multilayer-assembled nanovaccine successfully integrated antigens and multiple agonists to modulate APC activation. A DNA hydrogel contributed to the control of APC's immune behaviors, including cell recruitment, activation, and migration, and induced robust antitumor responses as an all-in-one designable platform. In this Account, by summarizing strategies for both T-cell- and B-cell-based vaccine design, we not only compare the differences but also address the intrinsic uniformity between such vaccine designs and further discuss the potential of a combined T-cell- and B-cell-based vaccine, which highlights the applicability and feasibility of chemical strategies.
Collapse
Affiliation(s)
- Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing 100084, China
| | - Jing-Yun Su
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing 100084, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing 100084, China.,Beijing Institute for Brain Disorders, 10 Youanmenwai Xitoutiao, Fengtai District, Beijing 100069, China.,Center for Synthetic and Systems Biology, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing 100084, China
| |
Collapse
|
5
|
Li Z, Yang D, Guo T, Lin M. Advances in MUC1-Mediated Breast Cancer Immunotherapy. Biomolecules 2022; 12:biom12070952. [PMID: 35883508 PMCID: PMC9313386 DOI: 10.3390/biom12070952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
Breast cancer (BRCA) is the leading cause of death from malignant tumors among women. Fortunately, however, immunotherapy has recently become a prospective BRCA treatment with encouraging achievements and mild safety profiles. Since the overexpression and aberrant glycosylation of MUC1 (human mucin) are closely associated with BRCA, it has become an ideal target for BRCA immunotherapies. In this review, the structure and function of MUC1 are briefly introduced, and the main research achievements in different kinds of MUC1-mediated BRCA immunotherapy are highlighted, from the laboratory to the clinic. Afterward, the future directions of MUC1-mediated BRCA immunotherapy are predicted, addressing, for example, urgent issues in regard to how efficient immunotherapeutic strategies can be generated.
Collapse
Affiliation(s)
- Zhifeng Li
- Medical School of Nantong University, Nantong 226019, China; (Z.L.); (D.Y.)
| | - Dazhuang Yang
- Medical School of Nantong University, Nantong 226019, China; (Z.L.); (D.Y.)
| | - Ting Guo
- Research Center of Clinical Medicine, Jiangsu Taizhou People’s Hospital (Affiliated Hospital 5 of Nantong University), Taizhou 225300, China;
| | - Mei Lin
- Research Center of Clinical Medicine, Jiangsu Taizhou People’s Hospital (Affiliated Hospital 5 of Nantong University), Taizhou 225300, China;
- Correspondence:
| |
Collapse
|
6
|
Panasiuk M, Zimmer K, Czarnota A, Narajczyk M, Peszyńska-Sularz G, Chraniuk M, Hovhannisyan L, Żołędowska S, Nidzworski D, Żaczek AJ, Gromadzka B. Chimeric virus-like particles presenting tumour-associated MUC1 epitope result in high titers of specific IgG antibodies in the presence of squalene oil-in-water adjuvant: towards safe cancer immunotherapy. J Nanobiotechnology 2022; 20:160. [PMID: 35351156 PMCID: PMC8961490 DOI: 10.1186/s12951-022-01357-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background Immunotherapy is emerging as a powerful treatment approach for several types of cancers. Modulating the immune system to specifically target cancer cells while sparing healthy cells, is a very promising approach for safer therapies and increased survival of cancer patients. Tumour-associated antigens are favorable targets for cancer immunotherapy, as they are exclusively expressed by the cancer cells, minimizing the risk of an autoimmune reaction. The ability to initiate the activation of the immune system can be achieved by virus-like particles (VLPs) which are safe and potent delivery tools. VLP‐based vaccines have evolved dramatically over the last few decades and showed great potential in preventing infectious diseases. Immunogenic potency of engineered VLPs as a platform for the development of effective therapeutic cancer vaccines has been studied extensively. This study involves recombinant VLPs presenting multiple copies of tumour-specific mucin 1 (MUC1) epitope as a potentially powerful tool for future immunotherapy. Results In this report VLPs based on the structural protein of Norovirus (NoV VP1) were genetically modified to present multiple copies of tumour-specific MUC1 epitope on their surface. Chimeric MUC1 particles were produced in the eukaryotic Leishmania tarentolae expression system and used in combination with squalene oil-in-water emulsion MF59 adjuvant to immunize BALB/c mice. Sera from vaccinated mice demonstrated high titers of IgG and IgM antibodies which were specifically recognizing MUC1 antigen. Conclusions The obtained results show that immunization with recombinant chimeric NoV VP1- MUC1 VLPs result in high titers of MUC1 specific IgG antibodies and show great therapeutic potential as a platform to present tumour-associated antigens. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Mirosława Panasiuk
- Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, A. Abrahama 58, 80-307, Gdańsk, Poland.,NanoExpo Sp. z o.o., Kładki 24, 80-822, Gdańsk, Poland.,Department of in vitro Studies, Institute of Biotechnology and Molecular Medicine, Kampinoska 25, 80-180, Gdańsk, Poland
| | - Karolina Zimmer
- Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, A. Abrahama 58, 80-307, Gdańsk, Poland.,Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa 2, 43-309, Bielsko-Biala, Poland
| | - Anna Czarnota
- Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, A. Abrahama 58, 80-307, Gdańsk, Poland
| | - Magdalena Narajczyk
- Laboratory of Electron Microscopy, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Grażyna Peszyńska-Sularz
- Tri-City Central Animal Laboratory Research and Service Center, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| | - Milena Chraniuk
- Department of in vitro Studies, Institute of Biotechnology and Molecular Medicine, Kampinoska 25, 80-180, Gdańsk, Poland
| | - Lilit Hovhannisyan
- Department of in vitro Studies, Institute of Biotechnology and Molecular Medicine, Kampinoska 25, 80-180, Gdańsk, Poland
| | - Sabina Żołędowska
- Institute of Biotechnology and Molecular Medicine, Kampinoska 25, 80-180, Gdańsk, Poland
| | - Dawid Nidzworski
- Institute of Biotechnology and Molecular Medicine, Kampinoska 25, 80-180, Gdańsk, Poland
| | - Anna J Żaczek
- Laboratory of Translational Oncology, Medical University of Gdańsk, Dębinki 1, 80-210, Gdańsk, Poland
| | - Beata Gromadzka
- Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, A. Abrahama 58, 80-307, Gdańsk, Poland. .,NanoExpo Sp. z o.o., Kładki 24, 80-822, Gdańsk, Poland. .,Department of in vitro Studies, Institute of Biotechnology and Molecular Medicine, Kampinoska 25, 80-180, Gdańsk, Poland.
| |
Collapse
|
7
|
Roberto Raúl SG, Damaris IA, Ángel de Jesús JC, Leticia MF. Cry1Ac Protoxin Confers Antitumor Adjuvant Effect in a Triple-Negative Breast Cancer Mouse Model by Improving Tumor Immunity. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234211065154. [PMID: 35002244 PMCID: PMC8738886 DOI: 10.1177/11782234211065154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 11/17/2021] [Indexed: 12/07/2022] Open
Abstract
The Cry1Ac protoxin from Bacillus thuringiensis is a systemic
and mucosal adjuvant, able to confer protective immunity in different infection
murine models and induce both Th1 and TCD8+ cytotoxic lymphocyte responses,
which are required to induce antitumor immunity. The Cry1Ac toxin, despite
having not being characterized as an adjuvant, has also proved to be immunogenic
and able to activate macrophages. Here, we investigated the potential antitumor
adjuvant effect conferred by the Cry1Ac protoxin and Cry1Ac toxin in a triple
negative breast cancer (TNBC) murine model. First, we evaluated the ability of
Cry1Ac proteins to improve dendritic cell (DC) activation and cellular response
through intraperitoneal (i.p.) coadministration with the 4T1 cellular lysate.
Mice coadministered with the Cry1Ac protoxin showed an increase in the number
and activation of CD11c+MHCII- and CD11c+MHCII+low in the peritoneal
cavity and an increase in DC activation (CD11c+MHCII+) in the spleen. Cry1Ac
protoxin increased the proliferation of TCD4+ and TCD8+ lymphocytes in the
spleen and mesenteric lymph nodes (MLN), while the Cry1Ac toxin only increased
the proliferation of TCD4+ and TCD8+ in the MLN. Remarkably, when tested in the
in vivo TNBC mouse model, prophylactic immunizations with 4T1 lysates plus the
Cry1Ac protoxin protected mice from developing tumors. The antitumor effect
conferred by the Cry1Ac protoxin also increased specific cytotoxic T cell
responses, and prevented the typical tumor-related decrease of T cells
(TCD3+ and TCD4+) as well the increase of myeloid-derived suppressor cells
(MDSC) in spleen. Also in the tumor microenvironment of mice coadministered
twice with Cry1Ac protoxin immunological improvements were found such as
reductions in immunosupressive populations (T regulatory lymphocytes and MDSC)
along with increases in macrophages upregulating CD86. These results show a
differential antitumor adjuvant capability of Cry1Ac proteins, highlighting the
ability of Cry1Ac protoxin to enhance local and systemic tumor immunity in TNBC.
Finally, using a therapeutic approach, we evaluated the coadministration of
Cry1Ac protoxin with doxorubicin. A significant reduction in tumor volume and
lung metastasis was found, with increased intratumoral levels of tumor necrosis
factor-α and IL-6 with respect to the vehicle group, further supporting its
antitumor applicability.
Collapse
Affiliation(s)
- Servin-Garrido Roberto Raúl
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Ilhuicatzi-Alvarado Damaris
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Jiménez-Chávez Ángel de Jesús
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Moreno-Fierros Leticia
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| |
Collapse
|
8
|
Vafaei R, Samadi M, Hosseinzadeh A, Barzaman K, Esmailinejad M, Khaki Z, Farahmand L. Comparison of mucin-1 in human breast cancer and canine mammary gland tumor: a review study. Cancer Cell Int 2022; 22:14. [PMID: 35000604 PMCID: PMC8744232 DOI: 10.1186/s12935-021-02398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/08/2021] [Indexed: 11/10/2022] Open
Abstract
Mucin-1 (MUC-1) is a transmembrane glycoprotein, which bears many similarities between dogs and humans. Since the existence of animal models is essential to understand the significant factors involved in breast cancer mechanisms, canine mammary tumors (CMTs) could be used as a spontaneously occurring tumor model for human studies. Accordingly, this review assessed the comparison of canine and human MUC-1 based on their diagnostic and therapeutic aspects and showed how comparative oncology approaches could provide insights into translating pre-clinical trials from human to veterinary oncology and vice versa which could benefit both humans and dogs.
Collapse
Affiliation(s)
- Rana Vafaei
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, No.146, South Gandi Ave, Vanak Sq, Tehran, Iran
| | - Mitra Samadi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, No.146, South Gandi Ave, Vanak Sq, Tehran, Iran
| | - Aysooda Hosseinzadeh
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, No.146, South Gandi Ave, Vanak Sq, Tehran, Iran
| | - Khadijeh Barzaman
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, No.146, South Gandi Ave, Vanak Sq, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - MohammadReza Esmailinejad
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zohreh Khaki
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, No.146, South Gandi Ave, Vanak Sq, Tehran, Iran.
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, No.146, South Gandi Ave, Vanak Sq, Tehran, Iran.
| |
Collapse
|
9
|
Disis ML, Cecil DL. Breast cancer vaccines for treatment and prevention. Breast Cancer Res Treat 2021; 191:481-489. [PMID: 34846625 DOI: 10.1007/s10549-021-06459-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022]
Abstract
Breast cancer is immunogenic and a variety of vaccines have been designed to boost immunity directed against the disease. The components of a breast cancer vaccine, the antigen, the delivery system, and the adjuvant, can have a significant impact on vaccine immunogenicity. There have been numerous immunogenic proteins identified in all subtypes of breast cancer. The majority of these antigens are weakly immunogenic nonmutated tumor-associated proteins. Mutated proteins and neoantigen epitopes are found only in a small minority of patients and are enriched in the triple negative subtype. Several vaccines have advanced to large randomized Phase II or Phase III clinical trials. None of these trials met their primary endpoint of either progression-free or overall survival. Despite these set-backs investigators have learned important lessons regarding the clinical application of breast cancer vaccines from the type of immune response needed for tumor eradication, Type I T-cell immunity, to the patient populations most likely to benefit from vaccination. Many therapeutic breast cancer vaccines are now being tested in combination with other forms of immune therapy or chemotherapy and radiation. Breast cancer vaccines as single agents are now studied in the context of the prevention of relapse or development of disease. Newer approaches are designing vaccines to prevent breast cancer by intercepting high-risk lesions such as ductal carcinoma in situ to limit the progression of these tumors to invasive cancer. There are also several efforts to develop vaccines for the primary prevention of breast cancer by targeting antigens expressed during breast cancer initiation.
Collapse
Affiliation(s)
- Mary L Disis
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA.
| | - Denise L Cecil
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
10
|
Stergiou N, Urschbach M, Gabba A, Schmitt E, Kunz H, Besenius P. The Development of Vaccines from Synthetic Tumor-Associated Mucin Glycopeptides and their Glycosylation-Dependent Immune Response. CHEM REC 2021; 21:3313-3331. [PMID: 34812564 DOI: 10.1002/tcr.202100182] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022]
Abstract
Tumor-associated carbohydrate antigens are overexpressed as altered-self in most common epithelial cancers. Their glycosylation patterns differ from those of healthy cells, functioning as an ID for cancer cells. Scientists have been developing anti-cancer vaccines based on mucin glycopeptides, yet the interplay of delivery system, adjuvant and tumor associated MUC epitopes in the induced immune response is not well understood. The current state of the art suggests that the identity, abundancy and location of the glycans on the MUC backbone are all key parameters in the cellular and humoral response. This review shares lessons learned by us in over two decades of research in glycopeptide vaccines. By bridging synthetic chemistry and immunology, we discuss efforts in designing synthetic MUC1/4/16 vaccines and focus on the role of glycosylation patterns. We provide a brief introduction into the mechanisms of the immune system and aim to promote the development of cancer subunit vaccines.
Collapse
Affiliation(s)
- Natascha Stergiou
- Radionuclide Center, Radiology and Nuclear medicine Amsterdam UMC, VU University, De Boelelaan 1085c, 1081 HV, Amsterdam, the Netherlands
| | - Moritz Urschbach
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Adele Gabba
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Edgar Schmitt
- Institute of Immunology, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Horst Kunz
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Pol Besenius
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| |
Collapse
|
11
|
Liu Y, Li M, Zhu H, Jing Z, Yin X, Wang K, Hong Z, Zhao W. Alum colloid encapsulated inside β-glucan particles enhance humoral and CTL immune responses of MUC1 vaccine. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.01.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
12
|
Wu M, Wang S, Chen JY, Zhou LJ, Guo ZW, Li YH. Therapeutic cancer vaccine therapy for acute myeloid leukemia. Immunotherapy 2021; 13:863-877. [PMID: 33955237 DOI: 10.2217/imt-2020-0277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Antitumor function of the immune system has been harnessed to eradicate tumor cells as cancer therapy. Therapeutic cancer vaccines aim to help immune cells recognize tumor cells, which are difficult to target owing to immune escape. Many attempts at vaccine designs have been conducted throughout the last decades. In addition, as the advanced understanding of immunosuppressive mechanisms mediated by tumor cells, combining cancer vaccines with other immune therapies seems to be more efficient for cancer treatment. Acute myeloid leukemia (AML) is the most common acute leukemia in adults with poor prognosis. Evidence has shown T-cell-mediated immune responses in AML, which encourages the utility of immune therapies in AML. This review discusses cancer vaccines in AML from vaccine design as well as recent progress in vaccination combination with other immune therapies.
Collapse
Affiliation(s)
- Ming Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.,Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Sheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jian-Yu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Li-Juan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zi-Wen Guo
- Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Yu-Hua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
13
|
Gordon B, Gadi VK. The Role of the Tumor Microenvironment in Developing Successful Therapeutic and Secondary Prophylactic Breast Cancer Vaccines. Vaccines (Basel) 2020; 8:vaccines8030529. [PMID: 32937885 PMCID: PMC7565925 DOI: 10.3390/vaccines8030529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer affects roughly one in eight women over their lifetime and is a leading cause of cancer-related death in women. While outcomes have improved in recent years, prognosis remains poor for patients who present with either disseminated disease or aggressive molecular subtypes. Cancer immunotherapy has revolutionized the treatment of several cancers, with therapeutic vaccines aiming to direct the cytotoxic immune program against tumor cells showing particular promise. However, these results have yet to translate to breast cancer, which remains largely refractory from such approaches. Recent evidence suggests that the breast tumor microenvironment (TME) is an important and long understudied barrier to the efficacy of therapeutic vaccines. Through an improved understanding of the complex and biologically diverse breast TME, it may be possible to advance new combination strategies to render breast carcinomas sensitive to the effects of therapeutic vaccines. Here, we discuss past and present efforts to advance therapeutic vaccines in the treatment of breast cancer, the molecular mechanisms through which the TME contributes to the failure of such approaches, as well as the potential means through which these can be overcome.
Collapse
Affiliation(s)
- Benjamin Gordon
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL 60612, USA
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612, USA
- Correspondence:
| | - Vijayakrishna K. Gadi
- Division of Hematology and Oncology, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
14
|
Abstract
Personalized cancer vaccines (PCVs) are reinvigorating vaccine strategies in cancer immunotherapy. In contrast to adoptive T-cell therapy and checkpoint blockade, the PCV strategy modulates the innate and adaptive immune systems with broader activation to redeploy antitumor immunity with individualized tumor-specific antigens (neoantigens). Following a sequential scheme of tumor biopsy, mutation analysis, and epitope prediction, the administration of neoantigens with synthetic long peptide (SLP) or mRNA formulations dramatically improves the population and activity of antigen-specific CD4+ and CD8+ T cells. Despite the promising prospect of PCVs, there is still great potential for optimizing prevaccination procedures and vaccine potency. In particular, the arduous development of tumor-associated antigen (TAA)-based vaccines provides valuable experience and rational principles for augmenting vaccine potency which is expected to advance PCV through the design of adjuvants, delivery systems, and immunosuppressive tumor microenvironment (TME) reversion since current personalized vaccination simply admixes antigens with adjuvants. Considering the broader application of TAA-based vaccine design, these two strategies complement each other and can lead to both personalized and universal therapeutic methods. Chemical strategies provide vast opportunities for (1) exploring novel adjuvants, including synthetic molecules and materials with optimizable activity, (2) constructing efficient and precise delivery systems to avoid systemic diffusion, improve biosafety, target secondary lymphoid organs, and enhance antigen presentation, and (3) combining bioengineering methods to innovate improvements in conventional vaccination, "smartly" re-educate the TME, and modulate antitumor immunity. As chemical strategies have proven versatility, reliability, and universality in the design of T cell- and B cell-based antitumor vaccines, the union of such numerous chemical methods in vaccine construction is expected to provide new vigor and vitality in cancer treatment.
Collapse
Affiliation(s)
- Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China.,Beijing Institute for Brain Disorders, 100069 Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, 100084 Beijing, China
| |
Collapse
|
15
|
Danilenko ED, Belkina AO, Sysoeva GM. Development of Drugs Based on High-Polymeric Double-Stranded RNA for Antiviral and Antitumor Therapy. BIOCHEMISTRY (MOSCOW) SUPPLEMENT. SERIES B, BIOMEDICAL CHEMISTRY 2019; 13:308-323. [PMID: 32288939 PMCID: PMC7104317 DOI: 10.1134/s1990750819040036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 12/24/2022]
Abstract
Abstract-The review summarizes literature data on the development of drugs based on natural and synthetic high-polymeric double-stranded RNA (dsRNA), their antiviral, immunoadjuvant, and antitumor properties. Special attention is paid to cell receptors responding to exogenous dsRNA, pathways of dsRNA-dependent antiviral reaction, ability of dsRNA to inhibit growth and induce apoptosis of malignant cells. It has been shown that enhancing the innate immune response with dsRNA can be an effective component in improving methods for treating and preventing infectious and cancer diseases. The further use of dsRNA for the correction of pathological processes of different origin is discussed.
Collapse
Affiliation(s)
- E. D. Danilenko
- Institute of Medical Biotechnology, State Research Center of Virology and Biotechnology (SRC VB) “Vector”, Khimzavodskaya ul. 9, 633010 Berdsk, Novosibirsk region Russia
| | - A. O. Belkina
- Institute of Medical Biotechnology, State Research Center of Virology and Biotechnology (SRC VB) “Vector”, Khimzavodskaya ul. 9, 633010 Berdsk, Novosibirsk region Russia
| | - G. M. Sysoeva
- Institute of Medical Biotechnology, State Research Center of Virology and Biotechnology (SRC VB) “Vector”, Khimzavodskaya ul. 9, 633010 Berdsk, Novosibirsk region Russia
| |
Collapse
|
16
|
Danilenko ED, Belkina AO, Sysoeva GM. [Development of drugs on the basis of high-polymeric double-stranded RNA for antiviral and antitumor therapy]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:277-293. [PMID: 31436169 DOI: 10.18097/pbmc20196504277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review summarizes literature data on the development of drugs based on natural and synthetic high-polymeric double-stranded RNA, and their antiviral, immunoadjuvant and antitumor properties. Special attention is paid to cell receptors responding to exogenous dsRNA, the paths of dsRNA-dependent antiviral reaction, ability of dsRNA to inhibit growth and induce apoptosis ofmalignant cells. It has been shown that enhancing the innate immune response with dsRNA can be an effective component in improving methods for treating and preventing infectious and cancer diseases. The further use of dsRNA for the correction of pathological processes of different origin is discussed.
Collapse
Affiliation(s)
- E D Danilenko
- Institute of Medical Biotechnology, State Research Center of Virology and Biotechnology "Vector", Berdsk, Russia
| | - A O Belkina
- Institute of Medical Biotechnology, State Research Center of Virology and Biotechnology "Vector", Berdsk, Russia
| | - G M Sysoeva
- Institute of Medical Biotechnology, State Research Center of Virology and Biotechnology "Vector", Berdsk, Russia
| |
Collapse
|
17
|
Banerjee K, Gautam SK, Kshirsagar P, Ross KA, Spagnol G, Sorgen P, Wannemuehler MJ, Narasimhan B, Solheim JC, Kumar S, Batra SK, Jain M. Amphiphilic polyanhydride-based recombinant MUC4β-nanovaccine activates dendritic cells. Genes Cancer 2019; 10:52-62. [PMID: 31258832 PMCID: PMC6584211 DOI: 10.18632/genesandcancer.189] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mucin 4 (MUC4) is a high molecular weight glycoprotein that is differentially overexpressed in pancreatic cancer (PC), functionally contributes to disease progression, and correlates with poor survival. Further, due to its aberrant glycosylation and extensive splicing, MUC4 is a potential target for cancer immunotherapy. Our previous studies have demonstrated the utility of amphiphilic polyanhydride nanoparticles as a useful platform for the development of protein-based prophylactic and therapeutic vaccines. In the present study, we encapsulated purified recombinant human MUC4-beta (MUC4β) protein in polyanhydride (20:80 CPTEG:CPH) nanoparticles (MUC4β-nanovaccine) and evaluated its ability to activate dendritic cells and induce adaptive immunity. Immature dendritic cells when pulsed with MUC4β-nanovaccine exhibited significant increase in the surface expressions of MHC I and MHC II and costimulatory molecules (CD80 and CD86), as well as, secretion of pro-inflammatory cytokines (IFN-γ, IL-6, and IL-12) as compared to cells exposed to MUC4β alone or MUC4β mixed with blank nanoparticles (MUC4β+NP). Following immunization, as compared to the other formulations, MUC4β-nanovaccine elicited higher IgG2b to IgG1 ratio of anti-MUC4β-antibodies suggesting a predominantly Th1-like class switching. Thus, our findings demonstrate MUC4β-nanovaccine as a novel platform for PC immunotherapy.
Collapse
Affiliation(s)
- Kasturi Banerjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Prakash Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kathleen A Ross
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Gaelle Spagnol
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paul Sorgen
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA.,Nanovaccine Institute, Ames, IA and Omaha, NE, USA
| | - Balaji Narasimhan
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA.,Nanovaccine Institute, Ames, IA and Omaha, NE, USA
| | - Joyce C Solheim
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Nanovaccine Institute, Ames, IA and Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Nanovaccine Institute, Ames, IA and Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Nanovaccine Institute, Ames, IA and Omaha, NE, USA
| |
Collapse
|
18
|
Chen Y, Yuan F, Jiang X, Lv Q, Luo N, Gong C, Wang C, Yang L, He G. Discovery of a self-assembling and self-adjuvant lipopeptide as a saccharide-free peptide vaccine targeting EGFRvIII positive cutaneous melanoma. Biomater Sci 2018. [PMID: 29528348 DOI: 10.1039/c8bm00017d] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recently, tumor immunotherapy has achieved great progress in the treatment of hematological and solid neoplasms. The DC vaccines, KLH-conjugated vaccines or glycosylated peptide vaccines can efficiently induce immune responses against tumors. In the current study, we have discovered cholesteryl PADRE-EGFRvIII epitope-conjugated lipopeptide self-assembled micelles as a potential self-adjuvant vaccine against cutaneous melanoma. The lipopeptide vaccines were synthesized using a standard solid phase peptide synthesis method, and these vaccines could elicit both a humoral and a cellular immune response to EGFRvIII positive melanoma cells. Their high humoral immunoreaction stimulation properties in combination with their cytotoxic T-cell eliciting properties provide them with potent tumor inhibitory capacity. In therapeutic and preventive xenograft models of B16-EGFRvIII melanoma cells, the self-adjuvant lipopeptide vaccine micelles efficiently prevented tumor growth as well as tumorigenesis. Our results provide a novel platform for eliciting immune responses to non-antigenic cancer-related epitopes in peptide cancer vaccine discovery and development.
Collapse
Affiliation(s)
- Yujuan Chen
- State Key Laboratory of Biotherapy, Department of breast surgery and Department of dermatology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Li Q, Guo Z. Recent Advances in Toll Like Receptor-Targeting Glycoconjugate Vaccines. Molecules 2018; 23:molecules23071583. [PMID: 29966261 PMCID: PMC6100623 DOI: 10.3390/molecules23071583] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 02/04/2023] Open
Abstract
Many malignant cell surface carbohydrates resulting from abnormal glycosylation patterns of certain diseases can serve as antigens for the development of vaccines against these diseases. However, carbohydrate antigens are usually poorly immunogenic by themselves, thus they need to be covalently coupled with immunologically active carrier molecules to be functional. The most well established and commonly used carriers are proteins. In recent years, the use of toll-like receptor (TLR) ligands to formulate glycoconjugate vaccines has gained significant attention because TLR ligands can serve not only as carrier molecules but also as built-in adjuvants to form fully synthetic and self-adjuvanting conjugate vaccines, which have several advantages over carbohydrate-protein conjugates and formulated mixtures with external adjuvants. This article reviews recent progresses in the development of conjugate vaccines based on TLR ligands. Two major classes of TLR ligands, lipopeptides and lipid A derivatives will be covered with more focus on monophosohoryl lipid A (MPLA) and related analogs, which are TLR4 ligands demonstrated to be able to provoke T cell-dependent, adaptive immune responses. Corresponding conjugate vaccines have shown promising application potentials to multiple diseases including cancer.
Collapse
Affiliation(s)
- Qingjiang Li
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| |
Collapse
|
20
|
Vaccine therapy in hematologic malignancies. Blood 2018; 131:2640-2650. [DOI: 10.1182/blood-2017-11-785873] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/04/2018] [Indexed: 02/06/2023] Open
Abstract
Abstract
Immune-based therapy has emerged as a paradigm shift in cancer therapy with dramatic responses observed in previously incurable disease. Cancer vaccines are being developed to disrupt tumor-associated tolerance and activate and selectively expand tumor-specific lymphocytes within the native effector cell repertoire while maintaining immune-regulatory protection against autoimmunity. Although individual antigen approaches result in immune response with a suggestion of clinical effect in some settings, broader efficacy may be dependent on presentation of multiple antigens that capture clonal diversity presented in the context of functionally potent antigen-presenting cells. The use of whole cell–based strategies such as dendritic cell/tumor fusions have yielded provocative results in single-arm studies and are currently being explored in multicenter randomized trials. The posttransplant setting is a potentially promising platform for vaccination due to cytoreduction and relative depletion of inhibitory accessory cells fostering greater immune responsiveness. Integration of these efforts with other immunotherapeutic strategies and agents that target the tumor microenvironment is being studied in an effort to generate durable immunologic responses with clinically meaningful impact on disease.
Collapse
|
21
|
Glaffig M, Stergiou N, Hartmann S, Schmitt E, Kunz H. A Synthetic MUC1 Anticancer Vaccine Containing Mannose Ligands for Targeting Macrophages and Dendritic Cells. ChemMedChem 2017; 13:25-29. [PMID: 29193802 DOI: 10.1002/cmdc.201700646] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 11/27/2017] [Indexed: 01/28/2023]
Abstract
A MUC1 anticancer vaccine equipped with covalently linked divalent mannose ligands was found to improve the antigen uptake and presentation by targeting mannose-receptor-positive macrophages and dendritic cells. It induced much stronger specific IgG immune responses in mice than the non-mannosylated reference vaccine. Mannose coupling also led to increased numbers of macrophages, dendritic cells, and CD4+ T cells in the local lymph organs. Comparison of di- and tetravalent mannose ligands revealed an increased binding of the tetravalent version, suggesting that higher valency improves binding to the mannose receptor. The mannose-coupled vaccine and the non-mannosylated reference vaccine induced IgG antibodies that exhibited similar binding to human breast tumor cells.
Collapse
Affiliation(s)
- Markus Glaffig
- Johannes Gutenberg University Mainz, Institute of Organic Chemistry, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Natascha Stergiou
- Johannes Gutenberg University Mainz, University Medical Center Institute of Immunology, Langenbeckstraße 1, Building 708, 55131, Mainz, Germany
| | - Sebastian Hartmann
- Johannes Gutenberg University Mainz, Institute of Organic Chemistry, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Edgar Schmitt
- Johannes Gutenberg University Mainz, University Medical Center Institute of Immunology, Langenbeckstraße 1, Building 708, 55131, Mainz, Germany
| | - Horst Kunz
- Johannes Gutenberg University Mainz, Institute of Organic Chemistry, Duesbergweg 10-14, 55128, Mainz, Germany
| |
Collapse
|