1
|
Padzińska-Pruszyńska IB, Taciak B, Kiraga Ł, Smolarska A, Górczak M, Kucharzewska P, Kubiak M, Szeliga J, Matejuk A, Król M. Targeting Cancer: Microenvironment and Immunotherapy Innovations. Int J Mol Sci 2024; 25:13569. [PMID: 39769334 PMCID: PMC11679359 DOI: 10.3390/ijms252413569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
In 2024, the United States was projected to experience 2 million new cancer diagnoses and approximately 611,720 cancer-related deaths, reflecting a broader global trend in which cancer cases are anticipated to exceed 35 million by 2050. This increasing burden highlights ongoing challenges in cancer treatment despite significant advances that have reduced cancer mortality by 31% since 1991. Key obstacles include the disease's inherent heterogeneity and complexity, such as treatment resistance, cancer stem cells, and the multifaceted tumor microenvironment (TME). The TME-comprising various tumor and immune cells, blood vessels, and biochemical factors-plays a crucial role in tumor growth and resistance to therapies. Recent innovations in cancer treatment, particularly in the field of immuno-oncology, have leveraged insights into TME interactions. An emerging example is the FDA-approved therapy using tumor-infiltrating lymphocytes (TILs), demonstrating the potential of cell-based approaches in solid tumors. However, TIL therapy is just one of many strategies being explored. This review provides a comprehensive overview of the emerging field of immuno-oncology, focusing on how novel therapies targeting or harnessing components of the TME could enhance treatment efficacy and address persistent challenges in cancer care.
Collapse
Affiliation(s)
- Irena Barbara Padzińska-Pruszyńska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Bartłomiej Taciak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Łukasz Kiraga
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Anna Smolarska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Górczak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Kubiak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Jacek Szeliga
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, 65-046 Zielona Góra, Poland;
| | - Magdalena Król
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| |
Collapse
|
2
|
Martinis E, Ricci C, Trevisan C, Tomadini G, Tonon S. Cancer Vaccines: From the State of the Art to the Most Promising Frontiers in the Treatment of Colorectal Cancer. Pharmaceutics 2023; 15:1969. [PMID: 37514155 PMCID: PMC10383643 DOI: 10.3390/pharmaceutics15071969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Colorectal cancer represents 10% of all new cancer cases each year and accounts for almost 10% of all cancer deaths. According to the WHO, by 2040 there will be a 60% increase in colorectal cancer cases. These data highlight the need to explore new therapeutic strategies. Classical interventions include surgical resection, chemotherapy and radiotherapy, which are invasive strategies that have many side effects on the patients and greatly affect their quality of life. A great advance in the treatment of this cancer type, as well as of all the others, could be the development of a vaccination strategy preventing the onset, the progression or the relapse of the pathology. In this review, we summarize the main vaccination strategies that are being studied for the treatment of colorectal cancer (CRC) and finally explore the possibility of using B-cells for the development of a new type of vaccine.
Collapse
Affiliation(s)
- Eleonora Martinis
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Carolina Ricci
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Caterina Trevisan
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Gaia Tomadini
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Silvia Tonon
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| |
Collapse
|
3
|
Kim J, Thomas SN. Opportunities for Nitric Oxide in Potentiating Cancer Immunotherapy. Pharmacol Rev 2022; 74:1146-1175. [PMID: 36180108 PMCID: PMC9553106 DOI: 10.1124/pharmrev.121.000500] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/15/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
Despite nearly 30 years of development and recent highlights of nitric oxide (NO) donors and NO delivery systems in anticancer therapy, the limited understanding of exogenous NO's effects on the immune system has prevented their advancement into clinical use. In particular, the effects of exogenously delivered NO differing from that of endogenous NO has obscured how the potential and functions of NO in anticancer therapy may be estimated and exploited despite the accumulating evidence of NO's cancer therapy-potentiating effects on the immune system. After introducing their fundamentals and characteristics, this review discusses the current mechanistic understanding of NO donors and delivery systems in modulating the immunogenicity of cancer cells as well as the differentiation and functions of innate and adaptive immune cells. Lastly, the potential for the complex modulatory effects of NO with the immune system to be leveraged for therapeutic applications is discussed in the context of recent advancements in the implementation of NO delivery systems for anticancer immunotherapy applications. SIGNIFICANCE STATEMENT: Despite a 30-year history and recent highlights of nitric oxide (NO) donors and delivery systems as anticancer therapeutics, their clinical translation has been limited. Increasing evidence of the complex interactions between NO and the immune system has revealed both the potential and hurdles in their clinical translation. This review summarizes the effects of exogenous NO on cancer and immune cells in vitro and elaborates these effects in the context of recent reports exploiting NO delivery systems in vivo in cancer therapy applications.
Collapse
Affiliation(s)
- Jihoon Kim
- Parker H. Petit Institute for Bioengineering and Bioscience (J.K., S.N.T.), George W. Woodruff School of Mechanical Engineering (J.K., S.N.T.), and Wallace H. Coulter Department of Biomedical Engineering (S.N.T.), Georgia Institute of Technology, Atlanta, Georgia; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia (S.N.T.); and Division of Biological Science and Technology, Yonsei University, Wonju, South Korea (J.K.)
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience (J.K., S.N.T.), George W. Woodruff School of Mechanical Engineering (J.K., S.N.T.), and Wallace H. Coulter Department of Biomedical Engineering (S.N.T.), Georgia Institute of Technology, Atlanta, Georgia; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia (S.N.T.); and Division of Biological Science and Technology, Yonsei University, Wonju, South Korea (J.K.)
| |
Collapse
|
4
|
Xie Z, Ke Y, Chen J, Li Z, Wang C, Chen Y, Ding H, Cheng L. Prevalence and Spectrum of Predisposition Genes With Germline Mutations Among Chinese Patients With Bowel Cancer. Front Genet 2022; 12:755629. [PMID: 35154239 PMCID: PMC8829568 DOI: 10.3389/fgene.2021.755629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Bowel cancer is the third-most common cancer and the second leading cause of cancer-related death worldwide. Bowel cancer has a substantial hereditary component; however, additional hereditary risk factors involved in bowel cancer pathogenesis have not been systematically defined. Materials and Methods: A total of 573 patients with bowel cancer were enrolled in the present study, of whom 93.72% had colorectal cancer (CRC). Germline mutations were integrated with somatic mutation information via utilizing target next-generation sequencing. Results: Pathogenic/Likely Pathogenic (P/LP) germline alterations were identified in 47 (8.2%) patients with bowel cancer and the ratio of the number of these patients with family history was significantly higher in the P/LP group than that noted in the non-pathogenic (Non-P) group. Certain rare germline alterations were noted, such as those noted in the following genes: FANCD2, CDH1, and FLCN. A total of 32 patients (68.1%) had germline alterations in the DNA-damage repair (DDR) genes and homologous recombination (HR) accounted for the highest proportion of this subgroup. By comparing 573 patients with bowel cancer with reference controls (China_MAPs database), significant associations (p < 0.01) were observed between the incidence of bowel cancer and the presence of mutations in APC, ATM, MLH1, FANCD2, MSH3, MSH6, PMS1, and RAD51D. Somatic gene differential analysis revealed a marked difference in 18 genes and a significant difference was also noted in tumor mutation burden (TMB) between germline mutation carriers and non-germline mutation subjects (p < 0.001). In addition, TMB in DDR mutation groups indicated a dramatic difference compared with the non-DDR mutation group (p < 0.01). However, no statistically significant differences in TMB were noted among detailed DDR pathways for patients with bowel cancer, irrespective of the presence of germline mutations. Moreover, a significantly higher level (p < 0.0001) of mutation count was observed in the DDR group from The Cancer Genome Atlas (TCGA) database and the DDR and non-DDR alteration groups displayed various immune profiles. Conclusion: Chinese patients with bowel cancer exhibited a distinct spectrum of germline variants, with distinct molecular characteristics such as TMB and DDR. Furthermore, the information on somatic mutations obtained from TCGA database indicated that a deeper understanding of the interactions among DDR and immune cells would be useful to further investigate the role of DDR in bowel cancer.
Collapse
|
5
|
Page A, Hubert J, Fusil F, Cosset FL. Exploiting B Cell Transfer for Cancer Therapy: Engineered B Cells to Eradicate Tumors. Int J Mol Sci 2021; 22:9991. [PMID: 34576154 PMCID: PMC8468294 DOI: 10.3390/ijms22189991] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 01/22/2023] Open
Abstract
Nowadays, cancers still represent a significant health burden, accounting for around 10 million deaths per year, due to ageing populations and inefficient treatments for some refractory cancers. Immunotherapy strategies that modulate the patient's immune system have emerged as good treatment options. Among them, the adoptive transfer of B cells selected ex vivo showed promising results, with a reduction in tumor growth in several cancer mouse models, often associated with antitumoral immune responses. Aside from the benefits of their intrinsic properties, including antigen presentation, antibody secretion, homing and long-term persistence, B cells can be modified prior to reinfusion to increase their therapeutic role. For instance, B cells have been modified mainly to boost their immuno-stimulatory activation potential by forcing the expression of costimulatory ligands using defined culture conditions or gene insertion. Moreover, tumor-specific antigen presentation by infused B cells has been increased by ex vivo antigen loading (peptides, RNA, DNA, virus) or by the sorting/ engineering of B cells with a B cell receptor specific to tumor antigens. Editing of the BCR also rewires B cell specificity toward tumor antigens, and may trigger, upon antigen recognition, the secretion of antitumor antibodies by differentiated plasma cells that can then be recognized by other immune components or cells involved in tumor clearance by antibody-dependent cell cytotoxicity or complement-dependent cytotoxicity for example. With the expansion of gene editing methodologies, new strategies to reprogram immune cells with whole synthetic circuits are being explored: modified B cells can sense disease-specific biomarkers and, in response, trigger the expression of therapeutic molecules, such as molecules that counteract the tumoral immunosuppressive microenvironment. Such strategies remain in their infancy for implementation in B cells, but are likely to expand in the coming years.
Collapse
Affiliation(s)
| | | | | | - François-Loïc Cosset
- CIRI-Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 Allée d’Italie, F-69007 Lyon, France; (A.P.); (J.H.); (F.F.)
| |
Collapse
|
6
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 1006] [Impact Index Per Article: 251.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
7
|
Ren H, Zhang T, Wang Y, Yao Q, Wang Z, Zhang L, Wang L. Tumor-Derived Autophagosomes (DRibbles) Activate Human B Cells to Induce Efficient Antigen-Specific Human Memory T-Cell Responses. Front Immunol 2021; 12:675822. [PMID: 34122437 PMCID: PMC8187759 DOI: 10.3389/fimmu.2021.675822] [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: 03/04/2021] [Accepted: 05/07/2021] [Indexed: 11/13/2022] Open
Abstract
We have reported that tumor-derived autophagosomes (DRibbles) were efficient carriers of tumor antigens and DRibbles antigens could be present by DRibbles-activated B cells to stimulate effect and naïve T cells in mice. However, the effect of DRibbles on human B cells remains unclear. Herein, we found that DRibbles can also efficiently induce proliferation and activation of human B cells and lead to the production of chemokines, cytokines and hematopoietic growth factors. We further demonstrated human B cells can effectively phagocytose DRibbles directly and cross-present DRibbles antigens to stimulate antigen-specific memory T cells. Furthermore, we found that membrane-bound high-mobility group B1 (HMGB1) on DRibbles was crucial for inducing human B cells activation. Therefore, these findings provide further evidence to promote the clinical application of B-DRibbles vaccines.
Collapse
Affiliation(s)
- Hongyan Ren
- Department of Pathology and Pathophysiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianyu Zhang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, China
| | - Yongren Wang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Yao
- Department of Pathology and Pathophysiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziyu Wang
- Department of Pathology and Pathophysiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Luyao Zhang
- Department of Pathology and Pathophysiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lixin Wang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
8
|
Michaud D, Steward CR, Mirlekar B, Pylayeva-Gupta Y. Regulatory B cells in cancer. Immunol Rev 2021; 299:74-92. [PMID: 33368346 PMCID: PMC7965344 DOI: 10.1111/imr.12939] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022]
Abstract
Tumorigenesis proceeds through discrete steps where acquisition of genetic lesions and changes in the surrounding microenvironment combine to drive unrestricted neoplastic proliferation and metastasis. The ability of tumor-infiltrating immune cells to promote tumor growth via the provision of signals that enable tumor cell survival and proliferation as well as contribute to immune suppression is an active area of research. Recent efforts have provided us with mechanistic insights into how B cells can positively and negatively regulate immune responses. Negative regulation of immune responses in cancer can be mediated by regulatory B cells and is often a result of increased production of cytokines that can directly and indirectly affect anti-tumor immune function and cancer cell growth. Signals that lead to the expansion of regulatory B cells and the spectrum of their functional roles are not well understood and are the subject of active research by many groups. Here, we elaborate broadly on the history of regulatory B cells in cancer and summarize recent studies that have established genetic models for the study of regulatory B cell function and their potential for therapeutic intervention in the setting of solid cancers.
Collapse
Affiliation(s)
- Daniel Michaud
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Colleen R Steward
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
9
|
Rubio AJ, Porter T, Zhong X. Duality of B Cell-CXCL13 Axis in Tumor Immunology. Front Immunol 2020; 11:521110. [PMID: 33193299 PMCID: PMC7609404 DOI: 10.3389/fimmu.2020.521110] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor immunity is a rapidly evolving area of research consisting of many possible permutations of immune cell tumor interactions that are dependent upon cell type, tumor type, and stage in tumor progression. At the same time, the majority of cancer immunotherapies have been focused on modulating the T cell-mediated antitumor immune response and have largely ignored the potential utility that B cells possess with respect to tumor immunity. Therefore, this motivated an exploration into the role that B cells and their accompanying chemokine, CXCL13, play in tumor immunity across multiple tumor types. Both B cells and CXCL13 possess dualistic impacts on tumor progression and tumor immunity which is furthered detail in this review. Specifically, various B cells subtypes are able to suppress or enhance several important immunological functions. Paradoxically, CXCL13 has been shown to drive several pro-growth and invasive signaling pathways across multiple tumor types, while also, correlating with improved survival and immune cell tumor localization in other tumor types. Potential tools for better elucidating the mechanisms by which B cells and CXCL13 impact the antitumor immune response are also discussed. In addition, multiples strategies are proposed for modulating the B cell-CXCL13 axis for cancer immunotherapies.
Collapse
Affiliation(s)
- Angel J. Rubio
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States
| | - Tyrone Porter
- Department of Biomedical Engineering, University of Texas Austin, Austin, TX, United States
| | - Xuemei Zhong
- Hematology and Medical Oncology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
10
|
Mi JL, Xu M, Liu C, Wang RS. Interactions between tumor mutation burden and immune infiltration in ovarian cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2513-2523. [PMID: 33165430 PMCID: PMC7642696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/14/2020] [Indexed: 06/11/2023]
Abstract
The aim of the study was to evaluate the relationship between tumor mutational burden (TMB) and immune infiltration in ovarian cancer. We extracted somatic mutational data and gene expression profiles of ovarian cancer from The Cancer Genome Atlas (TCGA). The samples were separated into low and high TMB groups. Correlations between TMB and cancer prognosis were analyzed and immune cell infiltration in the high and low TMB subgroups was calculated using the CIBERSORT package software. High TMB was significantly related to an improved survival rate. We identified 4 TMB-related core genes that were significantly associated with prognosis. Furthermore, mutations in the 4 genes were associated with immune cell infiltration. We also found a high proportion of naive B cells and activated NK cells in the high TMB group, while increased proportions of memory B cells and plasma cells were found in the low TMB group. Overall, our study indicated that patients with a higher TMB level experienced a favorable survival outcome and this may influence immune infiltration in ovarian cancer. Furthermore, the 4 TMB-related core genes were highly correlated with prognosis and the level of immune cell infiltration.
Collapse
Affiliation(s)
- Jing-Lin Mi
- Department of Radiotherapy Oncology Clinical Medical Research Center, Guangxi Medical UniversityNanning, People’s Republic of China
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, People’s Republic of China
| | - Meng Xu
- Department of Radiotherapy Oncology Clinical Medical Research Center, Guangxi Medical UniversityNanning, People’s Republic of China
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, People’s Republic of China
| | - Chang Liu
- Department of Radiotherapy Oncology Clinical Medical Research Center, Guangxi Medical UniversityNanning, People’s Republic of China
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, People’s Republic of China
| | - Ren-Sheng Wang
- Department of Radiotherapy Oncology Clinical Medical Research Center, Guangxi Medical UniversityNanning, People’s Republic of China
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, People’s Republic of China
| |
Collapse
|
11
|
Huang F, Zhao J, Wei Y, Wen Z, Zhang Y, Wang X, Shen Y, Wang LX, Pan N. Anti-Tumor Efficacy of an Adjuvant Built-In Nanovaccine Based on Ubiquitinated Proteins from Tumor Cells. Int J Nanomedicine 2020; 15:1021-1035. [PMID: 32103954 PMCID: PMC7025662 DOI: 10.2147/ijn.s237578] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/26/2020] [Indexed: 12/11/2022] Open
Abstract
Background and Aim We have previously identified ubiquitinated proteins (UPs) from tumor cell lysates as a promising vaccine for cancer immunotherapy in different mouse tumor models. In this study, we aimed at developing a highly efficient therapeutic adjuvant built-in nanovaccine (α-Al2O3-UPs) by a simple method, in which UPs from tumor cells could be efficiently and conveniently enriched by α-Al2O3 nanoparticles covalently coupled with Vx3 proteins (α-Al2O3-CONH-Vx3). Methods The α-Al2O3 nanoparticles were modified with 4-hydroxybenzoic acid followed by coupling with ubiquitin-binding protein Vx3. It was then used to enrich UPs from 4T1 cell lysate. The stability and the efficiency for the UPs enrichment of α-Al2O3-CONH-Vx3 were examined. The ability of α-Al2O3-UPs to activate DCs was examined in vitro subsequently. The splenocytes from the vaccinated mice were re-stimulated with inactivated tumor cells, and the IFN-γ secretion was detected by ELISA and flow cytometry. Moreover, the therapeutic efficacy of α-Al2O3-UPs, alone and in combination with chemotherapy, was examined in 4T1 tumor-bearing mice. Results Our results showed that α-Al2O3-UPs were successfully synthesized and abundant UPs from tumor cell lysate were enriched by the new method. In vitro study showed that compared to the physical mixture of α-Al2O3 nanoparticles and UPs (α-Al2O3+UPs), α-Al2O3-UPs stimulation resulted in higher upregulations of CD80, CD86, MHC class I, and MHC class II on DCs, indicating the higher ability of DC activation. Moreover, α-Al2O3-UPs elicited a more effective immune response in mice, demonstrated by higher IFN-γ secretion than α-Al2O3+UPs. Furthermore, α-Al2O3-UPs also exhibited a more potent effect on tumor growth inhibition and survival prolongation in 4T1 tumor-bearing mice. Notably, when in combination with low dose chemotherapy, the anti-tumor effect was further enhanced, rather than using α-Al2O3-UPs alone. Conclusion This study presents an adjuvant built-in nanovaccine generated by a new simple method that can be potentially applied to cancer immunotherapy and lays the experimental foundation for future clinical application.
Collapse
Affiliation(s)
- Fang Huang
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Jinjin Zhao
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Yiting Wei
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Zhifa Wen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Yue Zhang
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Xuru Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Yanfei Shen
- Department of Bioengineering, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Li-Xin Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Ning Pan
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| |
Collapse
|
12
|
Largeot A, Pagano G, Gonder S, Moussay E, Paggetti J. The B-side of Cancer Immunity: The Underrated Tune. Cells 2019; 8:cells8050449. [PMID: 31086070 PMCID: PMC6562515 DOI: 10.3390/cells8050449] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Tumor-infiltrating lymphocytes are known to be critical in controlling tumor progression. While the role of T lymphocytes has been extensively studied, the function of B cells in this context is still ill-defined. In this review, we propose to explore the role of B cells in tumor immunity. First of all we define their dual role in promoting and inhibiting cancer progression depending on their phenotype. To continue, we describe the influence of different tumor microenvironment factors such as hypoxia on B cells functions and differentiation. Finally, the role of B cells in response to therapy and as potential target is examined. In accordance with the importance of B cells in immuno-oncology, we conclude that more studies are required to throw light on the precise role of B cells in the tumor microenvironment in order to have a better understanding of their functions, and to design new strategies that efficiently target these cells by immunotherapy.
Collapse
Affiliation(s)
- Anne Largeot
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxenbourg, Luxembourg.
| | - Giulia Pagano
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxenbourg, Luxembourg.
| | - Susanne Gonder
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxenbourg, Luxembourg.
| | - Etienne Moussay
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxenbourg, Luxembourg.
| | - Jerome Paggetti
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxenbourg, Luxembourg.
| |
Collapse
|
13
|
Smith M, García-Martínez E, Pitter MR, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunology 2018; 7:e1526250. [PMID: 30524908 PMCID: PMC6279325 DOI: 10.1080/2162402x.2018.1526250] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Indexed: 12/14/2022] Open
Abstract
Toll-like receptor (TLR) agonists demonstrate therapeutic promise as immunological adjuvants for anticancer immunotherapy. To date, three TLR agonists have been approved by US regulatory agencies for use in cancer patients. Additionally, the potential of hitherto experimental TLR ligands to mediate clinically useful immunostimulatory effects has been extensively investigated over the past few years. Here, we summarize recent preclinical and clinical advances in the development of TLR agonists for cancer therapy.
Collapse
Affiliation(s)
- Melody Smith
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elena García-Martínez
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Michael R. Pitter
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- INSERM, U1015, Villejuif, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/ Paris V, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- INSERM, U1138, Paris, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/ Paris V, Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
14
|
Rossetti RAM, Lorenzi NPC, Yokochi K, Rosa MBSDF, Benevides L, Margarido PFR, Baracat EC, Carvalho JP, Villa LL, Lepique AP. B lymphocytes can be activated to act as antigen presenting cells to promote anti-tumor responses. PLoS One 2018; 13:e0199034. [PMID: 29975708 PMCID: PMC6033398 DOI: 10.1371/journal.pone.0199034] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 05/30/2018] [Indexed: 12/22/2022] Open
Abstract
Immune evasion by tumors includes several different mechanisms, including the inefficiency of antigen presenting cells (APCs) to trigger anti-tumor T cell responses. B lymphocytes may display a pro-tumoral role but can also be modulated to function as antigen presenting cells to T lymphocytes, capable of triggering anti-cancer immune responses. While dendritic cells, DCs, are the best APC population to activate naive T cells, DCs or their precursors, monocytes, are frequently modulated by tumors, displaying a tolerogenic phenotype in cancer patients. In patients with cervical cancer, we observed that monocyte derived DCs are tolerogenic, inhibiting allogeneic T cell activation compared to the same population obtained from patients with precursor lesions or cervicitis. In this work, we show that B lymphocytes from cervical cancer patients respond to treatment with sCD40L and IL-4 by increasing the CD80+CD86+ population, therefore potentially increasing their ability to activate T cells. To test if B lymphocytes could actually trigger anti-tumor T cell responses, we designed an experimental model where we harvested T and B lymphocytes, or dendritic cells, from tumor bearing donors, and after APC stimulation, transplanted them, together with T cells into RAG1-/- recipients, previously injected with tumor cells. We were able to show that anti-CD40 activated B lymphocytes could trigger secondary T cell responses, dependent on MHC-II expression. Moreover, we showed that dendritic cells were resistant to the anti-CD40 treatment and unable to stimulate anti-tumor responses. In summary, our results suggest that B lymphocytes may be used as a tool for immunotherapy against cancer.
Collapse
Affiliation(s)
| | | | - Kaori Yokochi
- Hospital Universitário, Universidade de São Paulo, São Paulo, Brazil
| | | | - Luciana Benevides
- Department of Immunology and Biochemistry, Faculdade de Medicina de Ribeirao Preto, University of Sao Paulo, Ribeirão Preto, Brazil
| | | | - Edmund Chada Baracat
- Hospital Universitário, Universidade de São Paulo, São Paulo, Brazil
- Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Jesus Paula Carvalho
- Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Luisa Lina Villa
- Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Instituto de Radiologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Paula Lepique
- Department of Immunology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Dong H, Wen ZF, Chen L, Zhou N, Liu H, Dong S, Hu HM, Mou Y. Polyethyleneimine modification of aluminum hydroxide nanoparticle enhances antigen transportation and cross-presentation of dendritic cells. Int J Nanomedicine 2018; 13:3353-3365. [PMID: 29922056 PMCID: PMC5995426 DOI: 10.2147/ijn.s164097] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background The aim of this study was to explore the feasibility of delivering tumor antigens and enhancing the antigen cross-presentation of dendritic cells (DCs) by aluminum hydroxide nanoparticle with polyethyleneimine (PEI) modification (LV@HPA/PEI). Materials and methods The LV@HPA nanoparticles were modified by PEI first, then the influence of LV@HPA/PEI on DCs was examined. The distinct expression of ovalbumin (OVA) protein transported into DCs by LV@HPA/PEI was observed by flow cytometry and Western blot. The biocompatibility of LV@HPA/PEI, maturity and antigen cross-presentation of DCs was observed in vitro. Tumor derived autophagosomes (DRibbles) combined with LV@HPA/PEI were loaded into DCs, and DC vaccines were used to immunize mice. The percentage of CD3+CD8+IFN-γ+ T cells in immunized mice was determined by flow cytometry. Additionally, the functional properties of the LV@HPA/PEI-DRibble-DCs vaccine were examined in vivo in PancO2 tumor-bearing mice. Results In our study, we described how LV@HPA/PEI can be a functionalized antigen delivery system with notable antigen transport effect and negligible cytotoxicity. It was found that LV@HPA/PEI could be easily internalized into DCs to assist antigen release into the cytoplasm. In addition, DCs matured gradually after loading with LV@HPA/PEI-OVA, which increased significantly the cytokine IL-12 secretion and expression of surface molecules CD80 and CD86. Interestingly, DCs loaded with LV@HPA/PEI-DRibbles could promote the activation of tumor-specific T cells both in murine and in human T cells. In the following in vivo experiments, the vaccine of LV@HPA/PEI-DRibble-DCs significantly inhibited tumor growth and improved the survival rate of the PancO2 tumor-bearing mice. Conclusion We established a high-performance anti-tumor vaccine of DCs loaded with LV@ HPA/PEI nanoparticles and tumor-associated antigens in autophagosomes (DRibbles), which could serve as a therapeutic strategy in cancer immunotherapy.
Collapse
Affiliation(s)
- Heng Dong
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Laboratory of Cancer Immunobiology, Robert W Franz Cancer Research Center, Earle A Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Zhi-Fa Wen
- Laboratory of Cancer Immunobiology, Robert W Franz Cancer Research Center, Earle A Chiles Research Institute, Providence Cancer Center, Portland, OR, USA.,Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Lin Chen
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Na Zhou
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hui Liu
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shiling Dong
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hong-Ming Hu
- Laboratory of Cancer Immunobiology, Robert W Franz Cancer Research Center, Earle A Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Yongbin Mou
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
16
|
Zhao J, Pan N, Huang F, Aldarouish M, Wen Z, Gao R, Zhang Y, Hu HM, Shen Y, Wang LX. Vx3-Functionalized Alumina Nanoparticles Assisted Enrichment of Ubiquitinated Proteins from Cancer Cells for Enhanced Cancer Immunotherapy. Bioconjug Chem 2018; 29:786-794. [PMID: 29382195 DOI: 10.1021/acs.bioconjchem.7b00578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A simple and effective strategy was developed to enrich ubiquitinated proteins (UPs) from cancer cell lysate using the α-Al2O3 nanoparticles covalently linked with ubiquitin binding protein (Vx3) (denoted as α-Al2O3-Vx3) via a chemical linker. The functionalized α-Al2O3-Vx3 showed long-term stability and high efficiency for the enrichment of UPs from cancer cell lysates. Flow cytometry analysis results indicated dendritic cells (DCs) could more effectively phagocytize the covalently linked α-Al2O3-Vx3-UPs than the physical mixture of α-Al2O3 and Vx3-UPs (α-Al2O3/Vx3-UPs). Laser confocal microscopy images revealed that α-Al2O3-Vx3-UPs localized within the autophagosome of DCs, which then cross-presented α-Al2O3-Vx3-UPs to CD8+ T cells in an autophagosome-related cross-presentation pathway. Furthermore, α-Al2O3-Vx3-UPs enhanced more potent antitumor immune response and antitumor efficacy than α-Al2O3/cell lysate or α-Al2O3/Vx3-UPs. This work highlights the potential of using the Vx3 covalently linked α-Al2O3 as a simple and effective platform to enrich UPs from cancer cells for the development of highly efficient therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Jinjin Zhao
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Ning Pan
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Fang Huang
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Mohanad Aldarouish
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Zhifa Wen
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Rong Gao
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Yuye Zhang
- School of Chemistry and Chemical Engineering , Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Hong-Ming Hu
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China.,Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute , Providence Portland Medical Center , Portland , Oregon 97213 United States
| | - Yanfei Shen
- Department of Bioengineering , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| | - Li-Xin Wang
- Department of Microbiology and Immunology , Medicine School of Southeast University , Nanjing , Jiangsu 210009 , P.R. China
| |
Collapse
|
17
|
Shin CA, Cho HW, Shin AR, Sohn HJ, Cho HI, Kim TG. Co-expression of CD40L with CD70 or OX40L increases B-cell viability and antitumor efficacy. Oncotarget 2018; 7:46173-46186. [PMID: 27323820 PMCID: PMC5216789 DOI: 10.18632/oncotarget.10068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/29/2016] [Indexed: 12/16/2022] Open
Abstract
Activated B-cells are a promising alternative source of antigen-presenting cells. They can generally be obtained in sufficient numbers for clinical use, but in most instances produce weak immune responses and therapeutic effects that are suboptimal for use in therapeutic cancer vaccines. To improve the immunogenic potency and therapeutic efficacy of B-cell-based vaccines, ex vivo-activated B-cells were transduced with recombinant lentiviruses in order to express additional costimulatory ligands—CD40L, CD70, OX40L, or 4-1BBL—either individually or in pairs (CD70/CD40L, OX40L/CD40L, or 4-1BBL/CD40L). We observed that the expression of CD40L molecules on B-cells was crucial for T-cell priming and activation. Administration of B-cells co-expressing CD40L with the other costimulatory ligands provided substantial antigen-specific CD8 T-cell responses capable of provoking in vivo proliferation and potent cytolytic activities. Notably, expression of CD40L augmented B-cell viability by inhibiting apoptosis through upregulated expression of the anti-apoptotic molecules BCL2, Bcl-xL and Bax. B-cells co-expressing CD40L with CD70, OX40L, or 4-1BBL induced potent therapeutic antitumor effects in a B16 melanoma model. Moreover, the combination of genetically-modified B-cell vaccines with programmed cell death-1 blockade potentiated the therapeutic efficacy. These results suggest that B-cells endowed with additional costimulatory ligands enable the design of effective vaccination strategies against cancer.
Collapse
Affiliation(s)
- Chang-Ae Shin
- Department of Microbiology and Immunology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Hyun-Woo Cho
- Department of Microbiology and Immunology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - A-Ri Shin
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Hyun-Jung Sohn
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Hyun-Il Cho
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Tai-Gyu Kim
- Department of Microbiology and Immunology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| |
Collapse
|
18
|
Münz C. Autophagy Proteins in Phagocyte Endocytosis and Exocytosis. Front Immunol 2017; 8:1183. [PMID: 29018446 PMCID: PMC5614925 DOI: 10.3389/fimmu.2017.01183] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023] Open
Abstract
Autophagy was initially described as a catabolic pathway that recycles nutrients of cytoplasmic constituents after lysosomal degradation during starvation. Since the immune system monitors products of lysosomal degradation via major histocompatibility complex (MHC) class II restricted antigen presentation, autophagy was found to process intracellular antigens for display on MHC class II molecules. In recent years, however, it has become apparent that the molecular machinery of autophagy serves phagocytes in many more membrane trafficking pathways, thereby regulating immunity to infectious disease agents. In this minireview, we will summarize the recent evidence that autophagy proteins regulate phagocyte endocytosis and exocytosis for myeloid cell activation, pathogen replication, and MHC class I and II restricted antigen presentation. Selective stimulation and inhibition of the respective functional modules of the autophagy machinery might constitute valid therapeutic options in the discussed disease settings.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
19
|
You L, Jin S, Zhu L, Qian W. Autophagy, autophagy-associated adaptive immune responses and its role in hematologic malignancies. Oncotarget 2017; 8:12374-12388. [PMID: 27902471 PMCID: PMC5355352 DOI: 10.18632/oncotarget.13583] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 11/11/2016] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a tightly regulated catabolic process that leads to the degradation of cytoplasmatic components such as aggregated/misfolded proteins and organelles through the lysosomal machinery. Recent studies suggest that autophagy plays such a role in the context of the anti-tumor immune response, make it an attractive target for cancer immunotherapy. Defective autophagy in hematopoietic stem cells may contribute to the development of hematologic malignancies, including leukemia, myelodysplastic syndrome, and lymphoproliferative disorder. In blood cancer cells, autophagy can either result in chemoresistance or induce autophagic cell death that may act as immunogenic. Based on the successful experimental findings in vitro and in vivo, clinical trials of autophagy inhibitor such as hydroxychloroquine in combination with chemotherapy in patients with blood cancers are currently underway. However, autophagy inactivation might impair autophagy-triggered anticancer immunity, whereas induction of autophagy might become an effective immunotherapy. These aspects are discussed in this review together with a brief introduction to the autophagic molecular machinery and its roles in hematologic malignancies.
Collapse
Affiliation(s)
- Liangshun You
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| | - Shenhe Jin
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| | - Li Zhu
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| | - Wenbin Qian
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| |
Collapse
|
20
|
Münz C. The Macroautophagy Machinery in Endo- and Exocytosis. J Mol Biol 2016; 429:473-485. [PMID: 27932293 DOI: 10.1016/j.jmb.2016.11.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/28/2016] [Accepted: 11/29/2016] [Indexed: 12/30/2022]
Abstract
Macroautophagy and the autophagy-related gene (Atg) machinery execute during their canonical function cytoplasmic constituent degradation in lysosomes. This canonical function contributes to pathogen restriction and intracellular antigen presentation on major histocompatibility complex (MHC) class II molecules to CD4+ T cells. However, in the recent years, it has become clear that the Atg machinery is also used for other membrane transport functions, including endocytosis and exocytosis. This review describes these non-canonical functions in the context of antigen presentation on MHC class I and II molecules to CD8+ and CD4+ T cells, respectively, and during viral replication. Future studies will need to address how the Atg machinery is modified for these non-canonical functions, gets recruited to the respective sites of membrane modification, and recruits alternative Atg interactors to execute endo- and exocytosis instead of macroautophagy.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, CH-8057, Switzerland.
| |
Collapse
|
21
|
Münz C. Autophagy Beyond Intracellular MHC Class II Antigen Presentation. Trends Immunol 2016; 37:755-763. [PMID: 27667710 DOI: 10.1016/j.it.2016.08.017] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 01/09/2023]
Abstract
Autophagy is a group of cellular pathways that deliver cytoplasmic constituents for lysosomal degradation. The peptides generated from these pathways can be presented by MHC II molecules, making autophagy an important source of antigens for CD4+ T cells. In addition, modules of the molecular machinery of autophagy were found in recent years to also influence extracellular antigen processing for MHC Class I and Class II presentation, as well as regulation of MHC Class I surface expression. These studies paint a more complicated picture of how regulation of individual autophagy proteins influences adaptive immunity. The respective pathways, especially in regard to their net outcome for CD4+ helper and CD8+ cytotoxic T cell responses in vivo, will be discussed in this review.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
22
|
Abstract
Autophagy describes catabolic pathways that deliver cytoplasmic constituents for lysosomal degradation. Since major histocompatibility complex (MHC) molecules sample protein degradation products and present them to T cells for adaptive immunity, it is maybe not too surprising that autophagy contributes to this protein antigen processing for MHC presentation. However, the recently recognized breath of pathways, by which autophagy contributes to MHC antigen processing, is exciting. Macroautophagy does not only seem to deliver intracellular but facilitates also extracellular antigen processing by lysosomal hydrolysis for MHC class II presentation. Moreover, even MHC class I molecules that usually display proteasomal products are regulated by macroautophagy, probably using a pool of these molecules outside the endoplasmic reticulum, where MHC class I molecules are loaded with peptide during canonical MHC class I antigen processing. This review aims to summarize these recent developments and point out gaps of knowledge, which should be filled by further investigation, in order to harness the different antigen-processing pathways via autophagy for vaccine improvement.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
From Antigen Delivery System to Adjuvanticy: The Board Application of Nanoparticles in Vaccinology. Vaccines (Basel) 2015; 3:930-9. [PMID: 26556378 PMCID: PMC4693225 DOI: 10.3390/vaccines3040930] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/11/2015] [Accepted: 10/30/2015] [Indexed: 11/17/2022] Open
Abstract
In the last years, nanotechnologies have raised great interest because of the potential applications of engineered nanoparticles in nanomedicine (i.e., in vaccination, in diagnostic imaging procedures, and as therapeutic drug delivery systems). The use of nanoparticles in medicine has brought about the issue of their interaction with the immune system for two main reasons: first, understanding how long nanomedicines could persist in the organism and exert their beneficial effects before being recognized and eliminated by our defensive systems; second, understanding how the immune responses can be modulated by nanoparticles in order to obtain optimal effects. This issue is crucial in vaccine formulations based on the use of nanoparticles, which can operate both as a delivery system to enhance antigen processing and as an immunostimulatory adjuvant to induce and amplify protective immunity, in part because of their ability to activate the inflammasome and induce the maturation of interleukin 1β. Nanoparticles can be excellent adjuvants due to their biocompatibility and their physicochemical properties (e.g., size, shape, and surface charge), which can be tailored to obtain different immunological effects. This review provides an overview of recent strategies for the use of nanoparticles as promising/attractive adjuvants for novel prophylactic and therapeutic vaccines. The use of nanovaccines, with their practically infinite possibilities of specific design, could open the way to precision vaccinology, i.e., vaccine formulations tailored on the individual immune reactivity status.
Collapse
|
24
|
Abstract
The central nervous system (CNS) possesses powerful local and global immunosuppressive capabilities that modulate unwanted inflammatory reactions in nervous tissue. These same immune-modulatory mechanisms are also co-opted by malignant brain tumors and pose a formidable challenge to brain tumor immunotherapy. Routes by which malignant gliomas coordinate immunosuppression include the mechanical and functional barriers of the CNS; immunosuppressive cytokines and catabolites; immune checkpoint molecules; tumor-infiltrating immune cells; and suppressor immune cells. The challenges to overcoming tumor-induced immunosuppression, however, are not unique to the brain, and several analogous immunosuppressive mechanisms also exist for primary tumors outside of the CNS. Ultimately, the immune responses in the CNS are linked and complementary to immune processes in the periphery, and advances in tumor immunotherapy in peripheral sites may therefore illuminate novel approaches to brain tumor immunotherapy, and vice versa.
Collapse
Affiliation(s)
- Powell Perng
- Department of Neurosurgery, School of Medicine, Johns Hopkins University , Baltimore, MD , USA
| | - Michael Lim
- Department of Neurosurgery, School of Medicine, Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|
25
|
Ubiquitinated proteins enriched from tumor cells by a ubiquitin binding protein Vx3(A7) as a potent cancer vaccine. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:34. [PMID: 25886865 PMCID: PMC4405905 DOI: 10.1186/s13046-015-0156-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/07/2015] [Indexed: 11/29/2022]
Abstract
Background Our previous studies have demonstrated that autophagosome-enriched vaccine (named DRibbles: DRiPs-containing blebs) induce a potent anti-tumor efficacy in different murine tumor models, in which DRibble-containing ubiquitinated proteins are efficient tumor-specific antigen source for the cross-presentation after being loaded onto dendritic cells. In this study, we sought to detect whether ubiquitinated proteins enriched from tumor cells could be used directly as a novel cancer vaccine. Methods The ubiquitin binding protein Vx3(A7) was used to isolate ubiquitinated proteins from EL4 and B16-F10 tumor cells after blocking their proteasomal degradation pathway. C57BL/6 mice were vaccinated with different doses of Ub-enriched proteins via inguinal lymph nodes or subcutaneous injection and with DRibbles, Ub-depleted proteins and whole cell lysate as comparison groups, respectively. The lymphocytes from the vaccinated mice were re-stimulated with inactivated tumor cells and the levels of IFN-γ in the supernatant were detected by ELISA. Anti-tumor efficacy of Ub-enriched proteins vaccine was evaluated by monitoring tumor growth in established tumor mice models. Graphpad Prism 5.0 was used for all statistical analysis. Results We found that after stimulation with inactivated tumor cells, the lymphocytes from the Ub-enriched proteins-vaccinated mice secreted high level of IFN-γ in dose dependent manner, in which the priming vaccination via inguinal lymph nodes injection induced higher IFN-γ level than that via subcutaneous injection. Moreover, the level of secreted IFN-γ in the Ub-enriched proteins group was markedly higher than that in the whole cell lysate and Ub-depleted proteins. Interestingly, the lymphocytes from mice vaccinated with Ub-enriched proteins, but not Ub-depleted proteins and whole cell lysates, isolated from EL4 or B16-F10 tumor cells also produced an obvious level of IFN-γ when stimulated alternately with inactivated B16-F10 or EL4 tumor cells. Furthermore, Ub-enriched proteins vaccine showed a significant inhibitory effect on in vivo growth of homologous tumor, as well as allogeneic tumor, compared with Ub-depleted proteins and tumor cell lysate. Tumor growth was regressed after three times of vaccination with Ub-enriched proteins in contrast to other groups. Conclusion These results indicated that Ub-enriched proteins isolated from tumor cells may have a potential as a potent vaccine for immunotherapy against cancer.
Collapse
|