1
|
Liu T, Yao W, Sun W, Yuan Y, Liu C, Liu X, Wang X, Jiang H. Components, Formulations, Deliveries, and Combinations of Tumor Vaccines. ACS NANO 2024; 18:18801-18833. [PMID: 38979917 DOI: 10.1021/acsnano.4c05065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Tumor vaccines, an important part of immunotherapy, prevent cancer or kill existing tumor cells by activating or restoring the body's own immune system. Currently, various formulations of tumor vaccines have been developed, including cell vaccines, tumor cell membrane vaccines, tumor DNA vaccines, tumor mRNA vaccines, tumor polypeptide vaccines, virus-vectored tumor vaccines, and tumor-in-situ vaccines. There are also multiple delivery systems for tumor vaccines, such as liposomes, cell membrane vesicles, viruses, exosomes, and emulsions. In addition, to decrease the risk of tumor immune escape and immune tolerance that may exist with a single tumor vaccine, combination therapy of tumor vaccines with radiotherapy, chemotherapy, immune checkpoint inhibitors, cytokines, CAR-T therapy, or photoimmunotherapy is an effective strategy. Given the critical role of tumor vaccines in immunotherapy, here, we look back to the history of tumor vaccines, and we discuss the antigens, adjuvants, formulations, delivery systems, mechanisms, combination therapy, and future directions of tumor vaccines.
Collapse
Affiliation(s)
- Tengfei Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyan Yao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yihan Yuan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Chen Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
2
|
Schweiger P, Hamann L, Strobel J, Weisbach V, Wandersee A, Christ J, Kehl S, Weidenthaler F, Antoniadis S, Hackstein H, Cunningham S. Functional Heterogeneity of Umbilical Cord Blood Monocyte-Derived Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:115-124. [PMID: 38809115 PMCID: PMC11215632 DOI: 10.4049/jimmunol.2400036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Human umbilical cord blood (UCB) represents a unique resource for hematopoietic stem cell transplantation for children and patients lacking suitable donors. UCB harbors a diverse set of leukocytes such as professional APCs, including monocytes, that could act as a novel source for cellular therapies. However, the immunological properties of UCB monocytes and monocyte-derived dendritic cells (MoDCs) are not fully characterized. In this study, we characterized the phenotype and functions of UCB-MoDCs to gauge their potential for future applications. UCB exhibited higher frequencies of platelets and lymphocytes as well as lower frequencies of neutrophils in comparison with adult whole blood. Leukocyte subset evaluation revealed significantly lower frequencies of granulocytes, NK cells, and CD14+CD16- monocytes. Surface marker evaluation revealed significantly lower rates of costimulatory molecules CD80 and CD83 while chemokine receptors CCR7 and CXCR4, as well as markers for Ag presentation, were similarly expressed. UCB-MoDCs were sensitive to TLR1-9 stimulation and presented quantitative differences in the release of proinflammatory cytokines. UCB-MoDCs presented functional CCR7-, CXCR4-, and CCR5-associated migratory behavior as well as adequate receptor- and micropinocytosis-mediated Ag uptake. When cocultured with allogeneic T lymphocytes, UCB-MoDCs induced weak CD4+ T lymphocyte proliferation, CD71 expression, and release of IFN-γ and IL-2. Taken together, UCB-MoDCs present potentially advantageous properties for future medical applications.
Collapse
Affiliation(s)
- Petra Schweiger
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Livia Hamann
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Julian Strobel
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Alexandra Wandersee
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Julia Christ
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Sven Kehl
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Filip Weidenthaler
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Sophia Antoniadis
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
3
|
Dang XTT, Phung CD, Lim CMH, Jayasinghe MK, Ang J, Tran T, Schwarz H, Le MTN. Dendritic cell-targeted delivery of antigens using extracellular vesicles for anti-cancer immunotherapy. Cell Prolif 2024; 57:e13622. [PMID: 38509634 PMCID: PMC11216926 DOI: 10.1111/cpr.13622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/25/2024] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
Neoantigen delivery using extracellular vesicles (EVs) has gained extensive interest in recent years. EVs derived from tumour cells or immune cells have been used to deliver tumour antigens or antitumor stimulation signals. However, potential DNA contamination from the host cell and the cost of large-scale EV production hinder their therapeutic applications in clinical settings. Here, we develop an antigen delivery platform for cancer vaccines from red blood cell-derived EVs (RBCEVs) targeting splenic DEC-205+ dendritic cells (DCs) to boost the antitumor effect. By loading ovalbumin (OVA) protein onto RBCEVs and delivering the protein to DCs, we were able to stimulate and present antigenic OVA peptide onto major histocompatibility complex (MHC) class I, subsequently priming activated antigen-reactive T cells. Importantly, targeted delivery of OVA using RBCEVs engineered with anti-DEC-205 antibody robustly enhanced antigen presentation of DCs and T cell activation. This platform is potentially useful for producing personalised cancer vaccines in clinical settings.
Collapse
Affiliation(s)
- Xuan T. T. Dang
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Cao Dai Phung
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Claudine Ming Hui Lim
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Migara Kavishka Jayasinghe
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Jorgen Ang
- School of Applied ScienceRepublic PolytechnicWoodlandsSingapore
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Infectious Disease Translational Research ProgramNational University of SingaporeSingaporeSingapore
- Immunology ProgrammeNational University of SingaporeSingaporeSingapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology ProgrammeNational University of SingaporeSingaporeSingapore
| | - Minh T. N. Le
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology ProgrammeNational University of SingaporeSingaporeSingapore
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute of Molecular and Cell BiologyAgency for Science, Technology, Technology and ResearchSingaporeSingapore
| |
Collapse
|
4
|
Aljieli M, Rivière C, Lantier L, Moiré N, Lakhrif Z, Boussemart AF, Cnudde T, Lajoie L, Aubrey N, Ahmed EM, Dimier-Poisson I, Di-Tommaso A, Mévélec MN. Specific Cell Targeting by Toxoplasma gondii Displaying Functional Single-Chain Variable Fragment as a Novel Strategy; A Proof of Principle. Cells 2024; 13:975. [PMID: 38891106 PMCID: PMC11172386 DOI: 10.3390/cells13110975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Toxoplasma gondii holds significant therapeutic potential; however, its nonspecific invasiveness results in off-target effects. The purpose of this study is to evaluate whether T. gondii specificity can be improved by surface display of scFv directed against dendritic cells' endocytic receptor, DEC205, and immune checkpoint PD-L1. Anti-DEC205 scFv was anchored to the T. gondii surface either directly via glycosylphosphatidylinositol (GPI) or by fusion with the SAG1 protein. Both constructs were successfully expressed, but the binding results suggested that the anti-DEC-SAG1 scFv had more reliable functionality towards recombinant DEC protein and DEC205-expressing MutuDC cells. Two anti-PD-L1 scFv constructs were developed that differed in the localization of the HA tag. Both constructs were adequately expressed, but the localization of the HA tag determined the functionality by binding to PD-L1 protein. Co-incubation of T. gondii displaying anti-PD-L1 scFv with tumor cells expressing/displaying different levels of PD-L1 showed strong binding depending on the level of available biomarker. Neutralization assays confirmed that binding was due to the specific interaction between anti-PD-L1 scFv and its ligand. A mixed-cell assay showed that T. gondii expressing anti-PD-L1 scFv predominately targets the PD-L1-positive cells, with negligible off-target binding. The recombinant RH-PD-L1-C strain showed increased killing ability on PD-L1+ tumor cell lines compared to the parental strain. Moreover, a co-culture assay of target tumor cells and effector CD8+ T cells showed that our model could inhibit PD1/PD-L1 interaction and potentiate T-cell immune response. These findings highlight surface display of antibody fragments as a promising strategy of targeting replicative T. gondii strains while minimizing nonspecific binding.
Collapse
Affiliation(s)
- Muna Aljieli
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
- Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Clément Rivière
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Louis Lantier
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Nathalie Moiré
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Zineb Lakhrif
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Anne-France Boussemart
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Thomas Cnudde
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Laurie Lajoie
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Nicolas Aubrey
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Elhadi M. Ahmed
- Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Isabelle Dimier-Poisson
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Anne Di-Tommaso
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| | - Marie-Noëlle Mévélec
- BioMAP, UMR ISP 1282 INRAE, Université de Tours, 37200 Tours, France; (M.A.); (C.R.); (L.L.); (N.M.); (Z.L.); (A.-F.B.); (L.L.); (N.A.); (I.D.-P.); (M.-N.M.)
| |
Collapse
|
5
|
Oláh I, Felföldi B, Benyeda Z, Magyar A, Nagy N, Soós Á, Szőcs E. Life cycle of chicken bursal secretory dendritic cell (BSDC). Poult Sci 2024; 103:103640. [PMID: 38688195 PMCID: PMC11077032 DOI: 10.1016/j.psj.2024.103640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 05/02/2024] Open
Abstract
The transmission electron microscopy revealed a dendritic cell in the medulla of the chicken bursal follicle. This dendritic cell has a classical secretory machinery; therefore, it has been named a bursal secretory dendritic cell (BSDC). The corticomedullary epithelial arch (CMEA) encloses lymphoid-like cells, which can proliferate and after entering the medulla, begin to differentiate to immature, then mature BSDC, which discharges glycoprotein (gp). With the exhaustion of gp production, the BSDC rapidly transforms into a macrophage-like cell (Mal), which is an activated endocytic cell of innate immunity. The Mal drifts through the follicle-associated epithelium (FAE)-supporting cells into the FAE, and via FAE, the Mal is eliminated in the bursal lumen. The infectious bursal disease virus (IBDV) infection accelerates the maturation process of BSDC precursors, which results in acute emptying of CMEA and subsequently, numerous immature BSDC(s) emerge. The IBDV infection stops the gp discharge, and the gp appears in the virus-containing Mal. The Movat pentachrome staining recognizes the gp in the extracellular spaces of the medulla and after infection in the Mal. The BSDC is the primary target of the IBDV. During IBDV infection, a large number of suddenly formed Mal actively migrate into the cortex, initiating cytokine storm and recruiting heterophil granulocytes. During embryogenesis, the vimentin-positive, possibly embryonic dendritic cells provide a microenvironment for carbohydrate switch. Around hatching, these embryonic, temporary dendritic cells get the Fc receptor, which bind maternal IgY. The posthatched forms of BSDC(s) gradually replace the embryonic ones and bind their own IgY.
Collapse
Affiliation(s)
- Imre Oláh
- Department of Anatomy, Histology and Embryology Semmelweis University 1094, Budapest, Hungary.
| | | | | | - Attila Magyar
- Department of Anatomy, Histology and Embryology Semmelweis University 1094, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology Semmelweis University 1094, Budapest, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology Semmelweis University 1094, Budapest, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology Semmelweis University 1094, Budapest, Hungary
| |
Collapse
|
6
|
Chop M, Ledo C, Nicolao MC, Loos J, Cumino A, Rodriguez Rodrigues C. Hydatid fluid from Echinococcus granulosus induces autophagy in dendritic cells and promotes polyfunctional T-cell responses. Front Cell Infect Microbiol 2024; 14:1334211. [PMID: 38817444 PMCID: PMC11137651 DOI: 10.3389/fcimb.2024.1334211] [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: 11/06/2023] [Accepted: 03/22/2024] [Indexed: 06/01/2024] Open
Abstract
Parasites possess remarkable abilities to evade and manipulate the immune response of their hosts. Echinococcus granulosus is a parasitic tapeworm that causes cystic echinococcosis in animals and humans. The hydatid fluid released by the parasite is known to contain various immunomodulatory components that manipulate host´s defense mechanism. In this study, we focused on understanding the effect of hydatid fluid on dendritic cells and its impact on autophagy induction and subsequent T cell responses. Initially, we observed a marked downregulation of two C-type lectin receptors in the cell membrane, CLEC9A and CD205 and an increase in lysosomal activity, suggesting an active cellular response to hydatid fluid. Subsequently, we visualized ultrastructural changes in stimulated dendritic cells, revealing the presence of macroautophagy, characterized by the formation of autophagosomes, phagophores, and phagolysosomes in the cell cytoplasm. To further elucidate the underlying molecular mechanisms involved in hydatid fluid-induced autophagy, we analyzed the expression of autophagy-related genes in stimulated dendritic cells. Our results demonstrated a significant upregulation of beclin-1, atg16l1 and atg12, indicating the induction of autophagy machinery in response to hydatid fluid exposure. Additionally, using confocal microscopy, we observed an accumulation of LC3 in dendritic cell autophagosomes, confirming the activation of this catabolic pathway associated with antigen presentation. Finally, to evaluate the functional consequences of hydatid fluid-induced autophagy in DCs, we evaluated cytokine transcription in the splenocytes. Remarkably, a robust polyfunctional T cell response, with inhibition of Th2 profile, is characterized by an increase in the expression of il-6, il-10, il-12, tnf-α, ifn-γ and tgf-β genes. These findings suggest that hydatid fluid-induced autophagy in dendritic cells plays a crucial role in shaping the subsequent T cell responses, which is important for a better understanding of host-parasite interactions in cystic echinococcosis.
Collapse
Affiliation(s)
- Maia Chop
- Instituto IQUIBIM, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Camila Ledo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
- Instituto IPROSAM, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Argentina
| | - María Celeste Nicolao
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
- Instituto IPROSAM, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Argentina
| | - Julia Loos
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
- Instituto IPROSAM, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Argentina
| | - Andrea Cumino
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
- Instituto IPROSAM, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Argentina
| | - Christian Rodriguez Rodrigues
- Instituto IQUIBIM, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| |
Collapse
|
7
|
Kong D, Qian Y, Yu B, Hu Z, Cheng C, Wang Y, Fang Z, Yu J, Xiang S, Cao L, He Y. Interaction of human dendritic cell receptor DEC205/CD205 with keratins. J Biol Chem 2024; 300:105699. [PMID: 38301891 PMCID: PMC10914487 DOI: 10.1016/j.jbc.2024.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/03/2024] Open
Abstract
DEC205 (CD205) is one of the major endocytic receptors on dendritic cells and has been widely used as a receptor target in immune therapies. It has been shown that DEC205 can recognize dead cells through keratins in a pH-dependent manner. However, the mechanism underlying the interaction between DEC205 and keratins remains unclear. Here we determine the crystal structures of an N-terminal fragment of human DEC205 (CysR∼CTLD3). The structural data show that DEC205 shares similar overall features with the other mannose receptor family members such as the mannose receptor and Endo180, but the individual domains of DEC205 in the crystal structure exhibit distinct structural features that may lead to specific ligand binding properties of the molecule. Among them, CTLD3 of DEC205 adopts a unique fold of CTLD, which may correlate with the binding of keratins. Furthermore, we examine the interaction of DEC205 with keratins by mutagenesis and biochemical assays based on the structural information and identify an XGGGX motif on keratins that can be recognized by DEC205, thereby providing insights into the interaction between DEC205 and keratins. Overall, these findings not only improve the understanding of the diverse ligand specificities of the mannose receptor family members at the molecular level but may also give clues for the interactions of keratins with their binding partners in the corresponding pathways.
Collapse
Affiliation(s)
- Dandan Kong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanying Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bowen Yu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Zhenzheng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Cheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Fang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Yu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Tianjin, China
| | - Song Xiang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Tianjin, China
| | - Longxing Cao
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Yongning He
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Lteif M, Pallardy M, Turbica I. Antibodies internalization mechanisms by dendritic cells and their role in therapeutic antibody immunogenicity. Eur J Immunol 2024; 54:e2250340. [PMID: 37985174 DOI: 10.1002/eji.202250340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Internalization and processing by antigen-presenting cells such as dendritic cells (DCs) are critical steps for initiating a T-cell response to therapeutic antibodies. Consequences are the production of neutralizing antidrug antibodies altering the clinical response, the presence of immune complexes, and, in some rare cases, hypersensitivity reactions. In recent years, significant progress has been made in the knowledge of cellular uptake mechanisms of antibodies in DCs. The uptake of antibodies could be directly related to their immunogenicity by regulating the quantity of materials entering the DCs in relation to antibody structure. Here, we summarize the latest insights into cellular uptake mechanisms and pathways in DCs. We highlight the approaches to study endocytosis, the impact of endocytosis routes on T-cell response, and discuss the link between how DCs internalize therapeutic antibodies and the potential mechanisms that could give rise to immunogenicity. Understanding these processes could help in developing assays to evaluate the immunogenicity potential of biotherapeutics.
Collapse
Affiliation(s)
- Maria Lteif
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Marc Pallardy
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Isabelle Turbica
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
9
|
Clemente B, Denis M, Silveira CP, Schiavetti F, Brazzoli M, Stranges D. Straight to the point: targeted mRNA-delivery to immune cells for improved vaccine design. Front Immunol 2023; 14:1294929. [PMID: 38090568 PMCID: PMC10711611 DOI: 10.3389/fimmu.2023.1294929] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
With the deepening of our understanding of adaptive immunity at the cellular and molecular level, targeting antigens directly to immune cells has proven to be a successful strategy to develop innovative and potent vaccines. Indeed, it offers the potential to increase vaccine potency and/or modulate immune response quality while reducing off-target effects. With mRNA-vaccines establishing themselves as a versatile technology for future applications, in the last years several approaches have been explored to target nanoparticles-enabled mRNA-delivery systems to immune cells, with a focus on dendritic cells. Dendritic cells (DCs) are the most potent antigen presenting cells and key mediators of B- and T-cell immunity, and therefore considered as an ideal target for cell-specific antigen delivery. Indeed, improved potency of DC-targeted vaccines has been proved in vitro and in vivo. This review discusses the potential specific targets for immune system-directed mRNA delivery, as well as the different targeting ligand classes and delivery systems used for this purpose.
Collapse
|
10
|
Wijfjes Z, van Dalen FJ, Le Gall CM, Verdoes M. Controlling Antigen Fate in Therapeutic Cancer Vaccines by Targeting Dendritic Cell Receptors. Mol Pharm 2023; 20:4826-4847. [PMID: 37721387 PMCID: PMC10548474 DOI: 10.1021/acs.molpharmaceut.3c00330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
Antigen-presenting cells (APCs) orchestrate immune responses and are therefore of interest for the targeted delivery of therapeutic vaccines. Dendritic cells (DCs) are professional APCs that excel in presentation of exogenous antigens toward CD4+ T helper cells, as well as cytotoxic CD8+ T cells. DCs are highly heterogeneous and can be divided into subpopulations that differ in abundance, function, and phenotype, such as differential expression of endocytic receptor molecules. It is firmly established that targeting antigens to DC receptors enhances the efficacy of therapeutic vaccines. While most studies emphasize the importance of targeting a specific DC subset, we argue that the differential intracellular routing downstream of the targeted receptors within the DC subset should also be considered. Here, we review the mouse and human receptors studied as target for therapeutic vaccines, focusing on antibody and ligand conjugates and how their targeting affects antigen presentation. We aim to delineate how targeting distinct receptors affects antigen presentation and vaccine efficacy, which will guide target selection for future therapeutic vaccine development.
Collapse
Affiliation(s)
- Zacharias Wijfjes
- Chemical
Immunology group, Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
- Institute
for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Floris J. van Dalen
- Chemical
Immunology group, Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
- Institute
for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Camille M. Le Gall
- Chemical
Immunology group, Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
- Institute
for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Martijn Verdoes
- Chemical
Immunology group, Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
- Institute
for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
11
|
Xiao Z, Wang R, Wang X, Yang H, Dong J, He X, Yang Y, Guo J, Cui J, Zhou Z. Impaired function of dendritic cells within the tumor microenvironment. Front Immunol 2023; 14:1213629. [PMID: 37441069 PMCID: PMC10333501 DOI: 10.3389/fimmu.2023.1213629] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Dendritic cells (DCs), a class of professional antigen-presenting cells, are considered key factors in the initiation and maintenance of anti-tumor immunity due to their powerful ability to present antigen and stimulate T-cell responses. The important role of DCs in controlling tumor growth and mediating potent anti-tumor immunity has been demonstrated in various cancer models. Accordingly, the infiltration of stimulatory DCs positively correlates with the prognosis and response to immunotherapy in a variety of solid tumors. However, accumulating evidence indicates that DCs exhibit a significantly dysfunctional state, ultimately leading to an impaired anti-tumor immune response due to the effects of the immunosuppressive tumor microenvironment (TME). Currently, numerous preclinical and clinical studies are exploring immunotherapeutic strategies to better control tumors by restoring or enhancing the activity of DCs in tumors, such as the popular DC-based vaccines. In this review, an overview of the role of DCs in controlling tumor progression is provided, followed by a summary of the current advances in understanding the mechanisms by which the TME affects the normal function of DCs, and concluding with a brief discussion of current strategies for DC-based tumor immunotherapy.
Collapse
Affiliation(s)
- Zhihua Xiao
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Ruiqi Wang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Xuyan Wang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Haikui Yang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Jiamei Dong
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Xin He
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Yang Yang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Jiahao Guo
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jiawen Cui
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhiling Zhou
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| |
Collapse
|
12
|
Leusmann S, Ménová P, Shanin E, Titz A, Rademacher C. Glycomimetics for the inhibition and modulation of lectins. Chem Soc Rev 2023; 52:3663-3740. [PMID: 37232696 PMCID: PMC10243309 DOI: 10.1039/d2cs00954d] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 05/27/2023]
Abstract
Carbohydrates are essential mediators of many processes in health and disease. They regulate self-/non-self- discrimination, are key elements of cellular communication, cancer, infection and inflammation, and determine protein folding, function and life-times. Moreover, they are integral to the cellular envelope for microorganisms and participate in biofilm formation. These diverse functions of carbohydrates are mediated by carbohydrate-binding proteins, lectins, and the more the knowledge about the biology of these proteins is advancing, the more interfering with carbohydrate recognition becomes a viable option for the development of novel therapeutics. In this respect, small molecules mimicking this recognition process become more and more available either as tools for fostering our basic understanding of glycobiology or as therapeutics. In this review, we outline the general design principles of glycomimetic inhibitors (Section 2). This section is then followed by highlighting three approaches to interfere with lectin function, i.e. with carbohydrate-derived glycomimetics (Section 3.1), novel glycomimetic scaffolds (Section 3.2) and allosteric modulators (Section 3.3). We summarize recent advances in design and application of glycomimetics for various classes of lectins of mammalian, viral and bacterial origin. Besides highlighting design principles in general, we showcase defined cases in which glycomimetics have been advanced to clinical trials or marketed. Additionally, emerging applications of glycomimetics for targeted protein degradation and targeted delivery purposes are reviewed in Section 4.
Collapse
Affiliation(s)
- Steffen Leusmann
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Petra Ménová
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Elena Shanin
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| |
Collapse
|
13
|
Becker J, Terracciano R, Yilmaz G, Napier R, Becer CR. Step-Growth Glycopolymers with a Defined Tacticity for Selective Carbohydrate-Lectin Recognition. Biomacromolecules 2023; 24:1924-1933. [PMID: 36976928 PMCID: PMC10091353 DOI: 10.1021/acs.biomac.3c00133] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Glycopolymers are potent candidates for biomedical applications by exploiting multivalent carbohydrate-lectin interactions. Owing to their specific recognition capabilities, glycosylated polymers can be utilized for targeted drug delivery to certain cell types bearing the corresponding lectin receptors. A fundamental challenge in glycopolymer research, however, is the specificity of recognition to receptors binding to the same sugar unit (e.g., mannose). Variation of polymer backbone chirality has emerged as an effective method to distinguish between lectins on a molecular level. Herein, we present a facile route toward producing glycopolymers with a defined tacticity based on a step-growth polymerization technique using click chemistry. A set of polymers have been fabricated and further functionalized with mannose moieties to enable lectin binding to receptors relevant to the immune system (mannose-binding lectin, dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin, and dendritic and thymic epithelial cell-205). Surface plasmon resonance spectrometry was employed to determine the kinetic parameters of the step-growth glycopolymers. The results highlight the importance of structural complexity in advancing glycopolymer synthesis, yet multivalency remains a main driving force in lectin recognition.
Collapse
Affiliation(s)
- Jonas Becker
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | | | - Gokhan Yilmaz
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | - Richard Napier
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K
| | - C Remzi Becer
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| |
Collapse
|
14
|
Achmad H, Saleh Ibrahim Y, Mohammed Al-Taee M, Gabr GA, Waheed Riaz M, Hamoud Alshahrani S, Alexis Ramírez-Coronel A, Turki Jalil A, Setia Budi H, Sawitri W, Elena Stanislavovna M, Gupta J. Nanovaccines in cancer immunotherapy: Focusing on dendritic cell targeting. Int Immunopharmacol 2022; 113:109434. [DOI: 10.1016/j.intimp.2022.109434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
|
15
|
Makandar AI, Jain M, Yuba E, Sethi G, Gupta RK. Canvassing Prospects of Glyco-Nanovaccines for Developing Cross-Presentation Mediated Anti-Tumor Immunotherapy. Vaccines (Basel) 2022; 10:vaccines10122049. [PMID: 36560459 PMCID: PMC9784904 DOI: 10.3390/vaccines10122049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
In view of the severe downsides of conventional cancer therapies, the quest of developing alternative strategies still remains of critical importance. In this regard, antigen cross-presentation, usually employed by dendritic cells (DCs), has been recognized as a potential solution to overcome the present impasse in anti-cancer therapeutic strategies. It has been established that an elevated cytotoxic T lymphocyte (CTL) response against cancer cells can be achieved by targeting receptors expressed on DCs with specific ligands. Glycans are known to serve as ligands for C-type lectin receptors (CLRs) expressed on DCs, and are also known to act as a tumor-associated antigen (TAA), and, thus, can be harnessed as a potential immunotherapeutic target. In this scenario, integrating the knowledge of cross-presentation and glycan-conjugated nanovaccines can help us to develop so called 'glyco-nanovaccines' (GNVs) for targeting DCs. Here, we briefly review and analyze the potential of GNVs as the next-generation anti-tumor immunotherapy. We have compared different antigen-presenting cells (APCs) for their ability to cross-present antigens and described the potential nanocarriers for tumor antigen cross-presentation. Further, we discuss the role of glycans in targeting of DCs, the immune response due to pathogens, and imitative approaches, along with parameters, strategies, and challenges involved in cross-presentation-based GNVs for cancer immunotherapy. It is known that the effectiveness of GNVs in eradicating tumors by inducing strong CTL response in the tumor microenvironment (TME) has been largely hindered by tumor glycosylation and the expression of different lectin receptors (such as galectins) by cancer cells. Tumor glycan signatures can be sensed by a variety of lectins expressed on immune cells and mediate the immune suppression which, in turn, facilitates immune evasion. Therefore, a sound understanding of the glycan language of cancer cells, and glycan-lectin interaction between the cancer cells and immune cells, would help in strategically designing the next-generation GNVs for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Amina I. Makandar
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Mannat Jain
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
- Correspondence: (E.Y.); (G.S.); or (R.K.G.)
| |
Collapse
|
16
|
Local immune cell contributions to fracture healing in aged individuals - A novel role for interleukin 22. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1262-1276. [PMID: 36028760 PMCID: PMC9440089 DOI: 10.1038/s12276-022-00834-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/25/2022] [Accepted: 06/06/2022] [Indexed: 11/08/2022]
Abstract
With increasing age, the risk of bone fractures increases while regenerative capacity decreases. This variation in healing potential appears to be linked to adaptive immunity, but the underlying mechanism is still unknown. This study sheds light on immunoaging/inflammaging, which impacts regenerative processes in aging individuals. In an aged preclinical model system, different levels of immunoaging were analyzed to identify key factors that connect immunoaged/inflammaged conditions with bone formation after long bone fracture. Immunological facets, progenitor cells, the microbiome, and confounders were monitored locally at the injury site and systemically in relation to healing outcomes in 12-month-old mice with distinct individual levels of immunoaging. Bone tissue formation during healing was delayed in the immunoaged group and could be associated with significant changes in cytokine levels. A prolonged and amplified pro-inflammatory reaction was caused by upregulated immune cell activation markers, increased chemokine receptor availability and a lack of inhibitory signaling. In immunoaged mice, interleukin-22 was identified as a core cell signaling protein that played a central role in delayed healing. Therapeutic neutralization of IL-22 reversed this specific immunoaging-related disturbed healing. Immunoaging was found to be an influencing factor of decreased regenerative capacity in aged individuals. Furthermore, a novel therapeutic strategy of neutralizing IL-22 may successfully rejuvenate healing in individuals with advanced immune experiences.
Collapse
|
17
|
Fresquet M, Lockhart-Cairns MP, Rhoden SJ, Jowitt TA, Briggs DC, Baldock C, Brenchley PE, Lennon R. Structure of PLA2R reveals presentation of the dominant membranous nephropathy epitope and an immunogenic patch. Proc Natl Acad Sci U S A 2022; 119:e2202209119. [PMID: 35858348 PMCID: PMC9303975 DOI: 10.1073/pnas.2202209119] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/31/2022] [Indexed: 01/04/2023] Open
Abstract
Membranous nephropathy is an autoimmune kidney disease caused by autoantibodies targeting antigens present on glomerular podocytes, instigating a cascade leading to glomerular injury. The most prevalent circulating autoantibodies in membranous nephropathy are against phospholipase A2 receptor (PLA2R), a cell surface receptor. The dominant epitope in PLA2R is located within the cysteine-rich domain, yet high-resolution structure-based mapping is lacking. In this study, we define the key nonredundant amino acids in the dominant epitope of PLA2R involved in autoantibody binding. We further describe two essential regions within the dominant epitope and spacer requirements for a synthetic peptide of the epitope for drug discovery. In addition, using cryo-electron microscopy, we have determined the high-resolution structure of PLA2R to 3.4 Å resolution, which shows that the dominant epitope and key residues within the cysteine-rich domain are accessible at the cell surface. In addition, the structure of PLA2R not only suggests a different orientation of domains but also implicates a unique immunogenic signature in PLA2R responsible for inducing autoantibody formation and recognition.
Collapse
Affiliation(s)
- Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Michael P. Lockhart-Cairns
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Samuel J. Rhoden
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Thomas A. Jowitt
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David C. Briggs
- Signalling and Structural Biology Lab, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Paul E. Brenchley
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
- Department of Paediatric Nephrology, Royal Manchester Children’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| |
Collapse
|
18
|
Nerb B, Dudziak D, Gessner A, Feuerer M, Ritter U. Have We Ignored Vector-Associated Microbiota While Characterizing the Function of Langerhans Cells in Experimental Cutaneous Leishmaniasis? FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.874081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
19
|
Structural and Functional Thymic Biomarkers Are Involved in the Pathogenesis of Thymic Epithelial Tumors: An Overview. IMMUNO 2022. [DOI: 10.3390/immuno2020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The normal human thymus originates from the third branchial cleft as two paired anlages that descend into the thorax and fuse on the midline of the anterior–superior mediastinum. Alongside the epithelial and lymphoid components, different types of lymphoid accessory cells, stromal mesenchymal and endothelial cells migrate to, or develop in, the thymus. After reaching maximum development during early postnatal life, the human thymus decreases in size and lymphocyte output drops with age. However, thymic immunological functions persist, although they deteriorate progressively. Several major techniques were fundamental to increasing the knowledge of thymic development and function during embryogenesis, postnatal and adult life; these include immunohistochemistry, immunofluorescence, flow cytometry, in vitro colony assays, transplantation in mice models, fetal organ cultures (FTOC), re-aggregated thymic organ cultures (RTOC), and whole-organ thymic scaffolds. The thymic morphological and functional characterization, first performed in the mouse, was then extended to humans. The purpose of this overview is to provide a report on selected structural and functional biomarkers of thymic epithelial cells (TEC) involved in thymus development and lymphoid cell maturation, and on the historical aspects of their characterization, with particular attention being paid to biomarkers also involved in Thymic Epithelial Tumor (TET) pathogenesis. Moreover, a short overview of targeted therapies in TET, based on currently available experimental and clinical data and on potential future advances will be proposed.
Collapse
|
20
|
Zhang H, Daněk O, Makarov D, Rádl S, Kim D, Ledvinka J, Vychodilová K, Hlaváč J, Lefèbre J, Denis M, Rademacher C, Ménová P. Drug-like Inhibitors of DC-SIGN Based on a Quinolone Scaffold. ACS Med Chem Lett 2022; 13:935-942. [DOI: 10.1021/acsmedchemlett.2c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/26/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Hengxi Zhang
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Ondřej Daněk
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Dmytro Makarov
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Stanislav Rádl
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
- Zentiva a.s., U Kabelovny 130, 10237 Prague 10, Czech Republic
| | - Dongyoon Kim
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Jiří Ledvinka
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Kristýna Vychodilová
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Jan Hlaváč
- Department of Organic Chemistry, Faculty of Science, Palacký University, Tř. 17. Listopadu 12, 77146 Olomouc, Czech Republic
| | - Jonathan Lefèbre
- Department of Pharmaceutical Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Maxime Denis
- Department of Pharmaceutical Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Petra Ménová
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| |
Collapse
|
21
|
Benne N, Ter Braake D, Stoppelenburg AJ, Broere F. Nanoparticles for Inducing Antigen-Specific T Cell Tolerance in Autoimmune Diseases. Front Immunol 2022; 13:864403. [PMID: 35392079 PMCID: PMC8981588 DOI: 10.3389/fimmu.2022.864403] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Autoimmune diseases affect many people worldwide. Current treatment modalities focus on the reduction of disease symptoms using anti-inflammatory drugs which can lead to side effects due to systemic immune suppression. Restoration of immune tolerance by down-regulating auto-reactive cells in an antigen-specific manner is currently the “holy grail” for the treatment of autoimmune diseases. A promising strategy is the use of nanoparticles that can deliver antigens to antigen-presenting cells which in turn can enhance antigen-specific regulatory T cells. In this review, we highlight some promising cell targets (e.g. liver sinusoidal endothelial cells and splenic marginal zone macrophages) for exploiting natural immune tolerance processes, and several strategies by which antigen-carrying nanoparticles can target these cells. We also discuss how nanoparticles carrying immunomodulators may be able to activate tolerance in other antigen-presenting cell types. Finally, we discuss some important aspects that must be taken into account when translating data from animal studies to patients.
Collapse
Affiliation(s)
- Naomi Benne
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Daniëlle Ter Braake
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Arie Jan Stoppelenburg
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Femke Broere
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
22
|
Small spleen peptides prevent development of psoriatic arthritis via restoration of peripheral tolerance. Mol Ther 2022; 30:745-762. [PMID: 34450252 PMCID: PMC8821926 DOI: 10.1016/j.ymthe.2021.08.030] [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: 02/23/2021] [Revised: 07/15/2021] [Accepted: 08/20/2021] [Indexed: 02/04/2023] Open
Abstract
The major challenge in the treatment of autoimmune diseases is the restoration of the impaired peripheral immune tolerance that always accompanies the development of such diseases. Here, we show that small splenic peptides (SSPs) of whole spleen extract efficiently suppress the development of psoriatic arthritis in vivo, even in the presence of sustained levels of pro-inflammatory cytokines. SSPs target dendritic cells (DCs) and convert them into tolerogenic cells, which in turn differentiate naive CD4+ cells into Foxp3-expressing T regulatory cells (Tregs). The latter requires direct contact between SSP-activated DCs and naive CD4+ T cells via PD-1 and CTLA4 immune checkpoint receptors of T cells. Finally, depletion of Foxp3+ Tregs in vivo abrogated the protective effect of SSPs on psoriatic arthritis development. We hypothesize that SSPs represent an intrinsic component of the adaptive immune system responsible for the physiological maintenance of peripheral tolerance and that therapeutically administered SSPs are able to restore imbalanced peripheral tolerance in autoimmune diseases.
Collapse
|
23
|
Bourque J, Hawiger D. Variegated Outcomes of T Cell Activation by Dendritic Cells in the Steady State. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:539-547. [PMID: 35042789 DOI: 10.4049/jimmunol.2100932] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022]
Abstract
Conventional dendritic cells (cDC) control adaptive immunity by sensing damage- and pathogen-associated molecular patterns and then inducing defined differentiation programs in T cells. Nevertheless, in the absence of specific proimmunogenic innate signals, generally referred to as the steady state, cDC also activate T cells to induce specific functional fates. Consistent with the maintenance of homeostasis, such specific outcomes of T cell activation in the steady state include T cell clonal anergy, deletion, and conversion of peripheral regulatory T cells (pTregs). However, the robust induction of protolerogenic mechanisms must be reconciled with the initiation of autoimmune responses and cancer immunosurveillance that are also observed under homeostatic conditions. Here we review the diversity of fates and functions of T cells involved in the opposing immunogenic and tolerogenic processes induced in the steady state by the relevant mechanisms of systemic cDC present in murine peripheral lymphoid organs.
Collapse
Affiliation(s)
- Jessica Bourque
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO
| |
Collapse
|
24
|
The Immunogenetics of Morphea and Lichen Sclerosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:155-172. [DOI: 10.1007/978-3-030-92616-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
25
|
Hiroki CH, Sarden N, Hassanabad MF, Yipp BG. Innate Receptors Expression by Lung Nociceptors: Impact on COVID-19 and Aging. Front Immunol 2021; 12:785355. [PMID: 34975876 PMCID: PMC8716370 DOI: 10.3389/fimmu.2021.785355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022] Open
Abstract
The lungs are constantly exposed to non-sterile air which carries harmful threats, such as particles and pathogens. Nonetheless, this organ is equipped with fast and efficient mechanisms to eliminate these threats from the airways as well as prevent pathogen invasion. The respiratory tract is densely innervated by sensory neurons, also known as nociceptors, which are responsible for the detection of external stimuli and initiation of physiological and immunological responses. Furthermore, expression of functional innate receptors by nociceptors have been reported; however, the influence of these receptors to the lung function and local immune response is poorly described. The COVID-19 pandemic has shown the importance of coordinated and competent pulmonary immunity for the prevention of pathogen spread as well as prevention of excessive tissue injury. New findings suggest that lung nociceptors can be a target of SARS-CoV-2 infection; what remains unclear is whether innate receptor trigger sensory neuron activation during SARS-CoV-2 infection and what is the relevance for the outcomes. Moreover, elderly individuals often present with respiratory, neurological and immunological dysfunction. Whether aging in the context of sensory nerve function and innate receptors contributes to the disorders of these systems is currently unknown. Here we discuss the expression of innate receptors by nociceptors, particularly in the lungs, and the possible impact of their activation on pulmonary immunity. We then demonstrate recent evidence that suggests lung sensory neurons as reservoirs for SARS-CoV-2 and possible viral recognition via innate receptors. Lastly, we explore the mechanisms by which lung nociceptors might contribute to disturbance in respiratory and immunological responses during the aging process.
Collapse
Affiliation(s)
- Carlos H. Hiroki
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nicole Sarden
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mortaza F. Hassanabad
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryan G. Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
26
|
Chitu V, Biundo F, Stanley ER. Colony stimulating factors in the nervous system. Semin Immunol 2021; 54:101511. [PMID: 34743926 DOI: 10.1016/j.smim.2021.101511] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Although traditionally seen as regulators of hematopoiesis, colony-stimulating factors (CSFs) have emerged as important players in the nervous system, both in health and disease. This review summarizes the cellular sources, patterns of expression and physiological roles of the macrophage (CSF-1, IL-34), granulocyte-macrophage (GM-CSF) and granulocyte (G-CSF) colony stimulating factors within the nervous system, with a particular focus on their actions on microglia. CSF-1 and IL-34, via the CSF-1R, are required for the development, proliferation and maintenance of essentially all CNS microglia in a temporal and regional specific manner. In contrast, in steady state, GM-CSF and G-CSF are mainly involved in regulation of microglial function. The alterations in expression of these growth factors and their receptors, that have been reported in several neurological diseases, are described and the outcomes of their therapeutic targeting in mouse models and humans are discussed.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
27
|
Phung CD, Tran TH, Nguyen HT, Nguyen TT, Jeong JH, Ku SK, Yong CS, Choi HG, Kim JO. Nanovaccines silencing IL-10 production at priming phase for boosting immune responses to melanoma. J Control Release 2021; 338:211-223. [PMID: 34419495 DOI: 10.1016/j.jconrel.2021.08.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 01/15/2023]
Abstract
Despite the significant efforts in developing cancer vaccines, there are still numerous challenges that need to be addressed to ensure their clinical efficacy. Herein, a lymphatic dendritic cell (DC)-targeted artificial nanovaccine mimicking tumor cell membrane (ATM-NV) is developed to boost effector immune response and control immunosuppression simultaneously. The NVs are formulated with lipids, tumor cell membrane proteins, imiquimod (IMQ), and IL-10 siRNA. IL-10 siRNA is incorporated to inhibit the secretion of IL-10, an immunosuppressive cytokine, of maturated DCs upon IMQ. To enhance the DC targeting ability, the nanovaccine surface was non-covalently conjugated with the anti-CD205 antibody. The IMQ and IL-10 siRNA co-loaded, CD205 receptor-targeted artificial tumor membrane NVs (IMQ/siR@ATM-NVs) efficiently migrate to the tumor-draining lymph node and target DCs. Furthermore, immunization with IMQ/siR@ATM-NVs reduces the production of IL-10 and increases Th1-driven antitumor immunity resulted in a great tumor inhibition efficacy. Our results suggest a potential strategy to promote the vaccination's antitumor efficacy by blocking the intrinsic negative regulators in DCs.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Tien Tiep Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Gyeonggi-do, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
28
|
Li J, Panetta F, O'Keeffe M, Leal Rojas IM, Radford KJ, Zhang JG, Fernandez-Ruiz D, Davey GM, Gully BS, Tullett KM, Rossjohn J, Berry R, Lee CN, Lahoud MH, Heath WR, Caminschi I. Elucidating the Motif for CpG Oligonucleotide Binding to the Dendritic Cell Receptor DEC-205 Leads to Improved Adjuvants for Liver-Resident Memory. THE JOURNAL OF IMMUNOLOGY 2021; 207:1836-1847. [PMID: 34479944 DOI: 10.4049/jimmunol.2001153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 11/19/2022]
Abstract
DEC-205 is a cell-surface receptor that transports bound ligands into the endocytic pathway for degradation or release within lysosomal endosomes. This receptor has been reported to bind a number of ligands, including keratin, and some classes of CpG oligodeoxynucleotides (ODN). In this study, we explore in detail the requirements for binding ODNs, revealing that DEC-205 efficiently binds single-stranded, phosphorothioated ODN of ≥14 bases, with preference for the DNA base thymidine, but with no requirement for a CpG motif. DEC-205 fails to bind double-stranded phosphodiester ODN, and thus does not bind the natural type of DNA found in mammals. The ODN binding preferences of DEC-205 result in strong binding of B class ODN, moderate binding to C class ODN, minimal binding to P class ODN, and no binding to A class ODN. Consistent with DEC-205 binding capacity, induction of serum IL-12p70 or activation of B cells by each class of ODN correlated with DEC-205 dependence in mice. Thus, the greater the DEC-205 binding capacity, the greater the dependence on DEC-205 for optimal responses. Finally, by covalently linking a B class ODN that efficiently binds DEC-205, to a P class ODN that shows poor binding, we improved DEC-205 binding and increased adjuvancy of the hybrid ODN. The hybrid ODN efficiently enhanced induction of effector CD8 T cells in a DEC-205-dependent manner. Furthermore, the hybrid ODN induced robust memory responses, and was particularly effective at promoting the development of liver tissue-resident memory T cells.
Collapse
Affiliation(s)
- Jessica Li
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Fatma Panetta
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Meredith O'Keeffe
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Ingrid M Leal Rojas
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kristen J Radford
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Jian-Guo Zhang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Gayle M Davey
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Benjamin S Gully
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - Kirsteen M Tullett
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Richard Berry
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - Chin-Nien Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Mireille H Lahoud
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - William R Heath
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia; .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Irina Caminschi
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| |
Collapse
|
29
|
Stephens AJ, Burgess-Brown NA, Jiang S. Beyond Just Peptide Antigens: The Complex World of Peptide-Based Cancer Vaccines. Front Immunol 2021; 12:696791. [PMID: 34276688 PMCID: PMC8279810 DOI: 10.3389/fimmu.2021.696791] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022] Open
Abstract
Peptide-based cancer vaccines rely upon the strong activation of the adaptive immune response to elicit its effector function. They have shown to be highly specific and safe, but have yet to prove themselves as an efficacious treatment for cancer in the clinic. This is for a variety of reasons, including tumour heterogeneity, self-tolerance, and immune suppression. Importance has been placed on the overall design of peptide-based cancer vaccines, which have evolved from simple peptide derivatives of a cancer antigen, to complex drugs; incorporating overlapping regions, conjugates, and delivery systems to target and stimulate different components of antigen presenting cells, and to bolster antigen cross-presentation. Peptide-based cancer vaccines are increasingly becoming more personalised to an individual's tumour antigen repertoire and are often combined with existing cancer treatments. This strategy ultimately aids in combating the shortcomings of a more generalised vaccine strategy and provides a comprehensive treatment, taking into consideration cancer cell variability and its ability to avoid immune interrogation.
Collapse
Affiliation(s)
- Alexander J Stephens
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Nicola A Burgess-Brown
- Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Shisong Jiang
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Barthlott T, Handel AE, Teh HY, Wirasinha RC, Hafen K, Žuklys S, Roch B, Orkin SH, de Villartay JP, Daley SR, Holländer GA. Indispensable epigenetic control of thymic epithelial cell development and function by polycomb repressive complex 2. Nat Commun 2021; 12:3933. [PMID: 34168132 PMCID: PMC8225857 DOI: 10.1038/s41467-021-24158-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 05/31/2021] [Indexed: 12/11/2022] Open
Abstract
Thymic T cell development and T cell receptor repertoire selection are dependent on essential molecular cues provided by thymic epithelial cells (TEC). TEC development and function are regulated by their epigenetic landscape, in which the repressive H3K27me3 epigenetic marks are catalyzed by polycomb repressive complex 2 (PRC2). Here we show that a TEC-targeted deficiency of PRC2 function results in a hypoplastic thymus with reduced ability to express antigens and select a normal repertoire of T cells. The absence of PRC2 activity reveals a transcriptomically distinct medullary TEC lineage that incompletely off-sets the shortage of canonically-derived medullary TEC whereas cortical TEC numbers remain unchanged. This alternative TEC development is associated with the generation of reduced TCR diversity. Hence, normal PRC2 activity and placement of H3K27me3 marks are required for TEC lineage differentiation and function and, in their absence, the thymus is unable to compensate for the loss of a normal TEC scaffold.
Collapse
Affiliation(s)
- Thomas Barthlott
- Department of Biomedicine and University Children's Hospital of Basel, University of Basel, Basel, Switzerland
| | - Adam E Handel
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Hong Ying Teh
- Department of Biomedicine and University Children's Hospital of Basel, University of Basel, Basel, Switzerland
| | - Rushika C Wirasinha
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Katrin Hafen
- Department of Biomedicine and University Children's Hospital of Basel, University of Basel, Basel, Switzerland
| | - Saulius Žuklys
- Department of Biomedicine and University Children's Hospital of Basel, University of Basel, Basel, Switzerland
| | - Benoit Roch
- Genome Dynamics in the Immune System Laboratory, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Stuart H Orkin
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Jean-Pierre de Villartay
- Genome Dynamics in the Immune System Laboratory, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Stephen R Daley
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- School of Health and Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Georg A Holländer
- Department of Biomedicine and University Children's Hospital of Basel, University of Basel, Basel, Switzerland.
- Department of Paediatrics and the Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
31
|
Franck CO, Fanslau L, Bistrovic Popov A, Tyagi P, Fruk L. Biopolymer-based Carriers for DNA Vaccine Design. Angew Chem Int Ed Engl 2021; 60:13225-13243. [PMID: 32893932 PMCID: PMC8247987 DOI: 10.1002/anie.202010282] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 12/16/2022]
Abstract
Over the last 30 years, genetically engineered DNA has been tested as novel vaccination strategy against various diseases, including human immunodeficiency virus (HIV), hepatitis B, several parasites, and cancers. However, the clinical breakthrough of the technique is confined by the low transfection efficacy and immunogenicity of the employed vaccines. Therefore, carrier materials were designed to prevent the rapid degradation and systemic clearance of DNA in the body. In this context, biopolymers are a particularly promising DNA vaccine carrier platform due to their beneficial biochemical and physical characteristics, including biocompatibility, stability, and low toxicity. This article reviews the applications, fabrication, and modification of biopolymers as carrier medium for genetic vaccines.
Collapse
Affiliation(s)
- Christoph O. Franck
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Luise Fanslau
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Andrea Bistrovic Popov
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Puneet Tyagi
- Dosage Form Design and DevelopmentBioPharmaceuticals DevelopmentR&DAstra ZenecaGaithersburgMD20878USA
| | - Ljiljana Fruk
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
32
|
Recent Progress in Dendritic Cell-Based Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13102495. [PMID: 34065346 PMCID: PMC8161242 DOI: 10.3390/cancers13102495] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Cancer immunotherapy has now attracted much attention because of the recent success of immune checkpoint inhibitors. However, they are only beneficial in a limited fraction of patients most probably due to lack of sufficient CD8+ cytotoxic T-lymphocytes against tumor antigens in the host. In this regard, dendritic cells are useful tools to induce host immune responses against exogenous antigens. In particular, recently characterized cross-presenting dendritic cells are capable of inducing CD8+ cytotoxic T-lymphocytes against exogenous antigens such as tumor antigens and uniquely express the chemokine receptor XCR1. Here we focus on the recent progress in DC-based cancer vaccines and especially the use of the XCR1 and its ligand XCL1 axis for the targeted delivery of cancer vaccines to cross-presenting dendritic cells. Abstract Cancer immunotherapy aims to treat cancer by enhancing cancer-specific host immune responses. Recently, cancer immunotherapy has been attracting much attention because of the successful clinical application of immune checkpoint inhibitors targeting the CTLA-4 and PD-1/PD-L1 pathways. However, although highly effective in some patients, immune checkpoint inhibitors are beneficial only in a limited fraction of patients, possibly because of the lack of enough cancer-specific immune cells, especially CD8+ cytotoxic T-lymphocytes (CTLs), in the host. On the other hand, studies on cancer vaccines, especially DC-based ones, have made significant progress in recent years. In particular, the identification and characterization of cross-presenting DCs have greatly advanced the strategy for the development of effective DC-based vaccines. In this review, we first summarize the surface markers and functional properties of the five major DC subsets. We then describe new approaches to induce antigen-specific CTLs by targeted delivery of antigens to cross-presenting DCs. In this context, the chemokine receptor XCR1 and its ligand XCL1, being selectively expressed by cross-presenting DCs and mainly produced by activated CD8+ T cells, respectively, provide highly promising molecular tools for this purpose. In the near future, CTL-inducing DC-based cancer vaccines may provide a new breakthrough in cancer immunotherapy alone or in combination with immune checkpoint inhibitors.
Collapse
|
33
|
Franck CO, Fanslau L, Bistrovic Popov A, Tyagi P, Fruk L. Biopolymer‐based Carriers for DNA Vaccine Design. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202010282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Christoph O. Franck
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Luise Fanslau
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Andrea Bistrovic Popov
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Puneet Tyagi
- Dosage Form Design and Development BioPharmaceuticals Development R&D Astra Zeneca Gaithersburg MD 20878 USA
| | - Ljiljana Fruk
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| |
Collapse
|
34
|
Gully BS, Venugopal H, Fulcher AJ, Fu Z, Li J, Deuss FA, Llerena C, Heath WR, Lahoud MH, Caminschi I, Rossjohn J, Berry R. The cryo-EM structure of the endocytic receptor DEC-205. J Biol Chem 2020; 296:100127. [PMID: 33257321 PMCID: PMC7948739 DOI: 10.1074/jbc.ra120.016451] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/23/2020] [Accepted: 11/30/2020] [Indexed: 11/06/2022] Open
Abstract
DEC-205 (CD205), a member of the macrophage mannose receptor protein family, is the prototypic endocytic receptor of dendritic cells, whose ligands include phosphorothioated cytosine-guanosine oligonucleotides, a motif often seen in bacterial or viral DNA. However, despite growing biological and clinical significance, little is known about the structural arrangement of this receptor or any of its family members. Here, we describe the 3.2 Å cryo-EM structure of human DEC-205, thereby illuminating the structure of the mannose receptor protein family. The DEC-205 monomer forms a compact structure comprising two intercalated rings of C-type lectin-like domains, where the N-terminal cysteine-rich and fibronectin domains reside at the central intersection. We establish a pH-dependent oligomerization pathway forming tetrameric DEC-205 using solution-based techniques and ultimately solved the 4.9 Å cryo-EM structure of the DEC-205 tetramer to identify the unfurling of the second lectin ring which enables tetramer formation. Furthermore, we suggest the relevance of this oligomerization pathway within a cellular setting, whereby cytosine-guanosine binding appeared to disrupt this cell-surface oligomer. Accordingly, we provide insight into the structure and oligomeric assembly of the DEC-205 receptor.
Collapse
Affiliation(s)
- Benjamin S Gully
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.
| | - Hariprasad Venugopal
- Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Victoria, Australia
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Clayton, Victoria, Australia
| | - Zhihui Fu
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jessica Li
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Felix A Deuss
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Carmen Llerena
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - William R Heath
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, Parkville, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Mireille H Lahoud
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Irina Caminschi
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom.
| | - Richard Berry
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
35
|
Abstract
After both sterile and infectious insults, damage is inflicted on tissues leading to accidental or programmed cell death. In addition, events of programmed cell death also take place under homeostatic conditions, such as in embryo development or in the turnover of hematopoietic cells. Mammalian tissues are seeded with myeloid immune cells, which harbor a plethora of receptors that allow the detection of cell death, modulating immune responses. The myeloid C-type lectin receptors (CLRs) are one of the most prominent families of receptors involved in tailoring immunity after sensing dead cells. In this chapter, we will cover a diversity of signals arising from different forms of cell death and how they are recognized by myeloid CLRs. We will also explore how myeloid cells develop their sentinel function, exploring how some of these CLRs identify cell death and the type of responses triggered thereof. In particular, we will focus on DNGR-1 (CLEC9A), Mincle (CLEC4E), CLL-1 (CLEC12A), LOX-1 (OLR1), CD301 (CLEC10A) and DEC-205 (LY75) as paradigmatic death-sensing CLRs expressed by myeloid cells. The molecular processes triggered after cell death recognition by myeloid CLRs contribute to the regulation of immune responses in pathologies associated with tissue damage, such as infection, autoimmunity and cancer. A better understanding of these processes may help to improve the current approaches for therapeutic intervention.
Collapse
|
36
|
Gaudio E, Tarantelli C, Spriano F, Guidetti F, Sartori G, Bordone R, Arribas AJ, Cascione L, Bigioni M, Merlino G, Fiascarelli A, Bressan A, Mensah AA, Golino G, Lucchini R, Bernasconi E, Rossi D, Zucca E, Stussi G, Stathis A, Boyd RS, Dusek RL, Bisht A, Attanasio N, Rohlff C, Pellacani A, Binaschi M, Bertoni F. Targeting CD205 with the antibody drug conjugate MEN1309/OBT076 is an active new therapeutic strategy in lymphoma models. Haematologica 2020; 105:2584-2591. [PMID: 33131247 PMCID: PMC7604571 DOI: 10.3324/haematol.2019.227215] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 01/02/2020] [Indexed: 11/22/2022] Open
Abstract
Antibody drug conjugates represent an important class of anti-cancer drugs in both solid tumors and hematological cancers. Here, we report preclinical data on the anti-tumor activity of the first-in-class antibody drug conjugate MEN1309/OBT076 targeting CD205. The study included preclinical in vitro activity screening on a large panel of cell lines, both as single agent and in combination and validation experiments on in vivo models. CD205 was first shown frequently expressed in lymphomas, leukemias and multiple myeloma by immunohistochemistry on tissue microarrays. Anti-tumor activity of MEN1309/OBT076 as single agent was then shown across 42 B-cell lymphoma cell lines with a median IC50 of 200 pM and induction of apoptosis in 25/42 (59.5%) of the cases. The activity appeared highly correlated with its target expression. After in vivo validation as the single agent, the antibody drug conjugate synergized with the BCL2 inhibitor venetoclax, and the anti-CD20 monoclonal antibody rituximab. The first-in-class antibody drug targeting CD205, MEN1309/OBT076, demonstrated strong pre-clinical anti-tumor activity in lymphoma, warranting further investigations as a single agent and in combination.
Collapse
Affiliation(s)
- Eugenio Gaudio
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Chiara Tarantelli
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Filippo Spriano
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Francesca Guidetti
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Giulio Sartori
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Roberta Bordone
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Alberto J. Arribas
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Luciano Cascione
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | | | | | | | | | - Afua Adjeiwaa Mensah
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Gaetanina Golino
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Renzo Lucchini
- Laboratorio di Diagnostica Molecolare, Dipartimento di Medicina di Laboratorio EOLAB, Bellinzona, Switzerland
| | - Elena Bernasconi
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
| | - Davide Rossi
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Emanuele Zucca
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Georg Stussi
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | | | | | | | | | | | | | | | | - Francesco Bertoni
- Universita della Svizzera italiana, Istituto Oncologico di Ricerca, Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| |
Collapse
|
37
|
The transcription factor C/EBPβ orchestrates dendritic cell maturation and functionality under homeostatic and malignant conditions. Proc Natl Acad Sci U S A 2020; 117:26328-26339. [PMID: 33020261 DOI: 10.1073/pnas.2008883117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Dendritic cell (DC) maturation is a prerequisite for the induction of adaptive immune responses against pathogens and cancer. Transcription factor (TF) networks control differential aspects of early DC progenitor versus late-stage DC cell fate decisions. Here, we identified the TF C/EBPβ as a key regulator for DC maturation and immunogenic functionality under homeostatic and lymphoma-transformed conditions. Upon cell-specific deletion of C/EBPβ in CD11c+MHCIIhi DCs, gene expression profiles of splenic C/EBPβ-/- DCs showed a down-regulation of E2F cell cycle target genes and associated proliferation signaling pathways, whereas maturation signatures were enriched. Total splenic DC cell numbers were modestly increased but differentiation into cDC1 and cDC2 subsets were unaltered. The splenic CD11c+MHCIIhiCD64+ DC compartment was also increased, suggesting that C/EBPβ deficiency favors the expansion of monocytic-derived DCs. Expression of C/EBPβ could be mimicked in LAP/LAP* isoform knockin DCs, whereas the short isoform LIP supported a differentiation program similar to deletion of the full-length TF. In accordance with E2F1 being a negative regulator of DC maturation, C/EBPβ-/- bone marrow-derived DCs matured much faster enabling them to activate and polarize T cells stronger. In contrast to a homeostatic condition, lymphoma-exposed DCs exhibited an up-regulation of the E2F transcriptional pathways and an impaired maturation. Pharmacological blockade of C/EBPβ/mTOR signaling in human DCs abrogated their protumorigenic function in primary B cell lymphoma cocultures. Thus, C/EBPβ plays a unique role in DC maturation and immunostimulatory functionality and emerges as a key factor of the tumor microenvironment that promotes lymphomagenesis.
Collapse
|
38
|
Huang MN, Nicholson LT, Batich KA, Swartz AM, Kopin D, Wellford S, Prabhakar VK, Woroniecka K, Nair SK, Fecci PE, Sampson JH, Gunn MD. Antigen-loaded monocyte administration induces potent therapeutic antitumor T cell responses. J Clin Invest 2020; 130:774-788. [PMID: 31661470 DOI: 10.1172/jci128267] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022] Open
Abstract
Efficacy of dendritic cell (DC) cancer vaccines is classically thought to depend on their antigen-presenting cell (APC) activity. Studies show, however, that DC vaccine priming of cytotoxic T lymphocytes (CTLs) requires the activity of endogenous DCs, suggesting that exogenous DCs stimulate antitumor immunity by transferring antigens (Ags) to endogenous DCs. Such Ag transfer functions are most commonly ascribed to monocytes, implying that undifferentiated monocytes would function equally well as a vaccine modality and need not be differentiated to DCs to be effective. Here, we used several murine cancer models to test the antitumor efficacy of undifferentiated monocytes loaded with protein or peptide Ag. Intravenously injected monocytes displayed antitumor activity superior to DC vaccines in several cancer models, including aggressive intracranial glioblastoma. Ag-loaded monocytes induced robust CTL responses via Ag transfer to splenic CD8+ DCs in a manner independent of monocyte APC activity. Ag transfer required cell-cell contact and the formation of connexin 43-containing gap junctions between monocytes and DCs. These findings demonstrate the existence of an efficient gap junction-mediated Ag transfer pathway between monocytes and CD8+ DCs and suggest that administration of tumor Ag-loaded undifferentiated monocytes may serve as a simple and efficacious immunotherapy for the treatment of human cancers.
Collapse
Affiliation(s)
- Min-Nung Huang
- Department of Immunology.,Division of Cardiology, Department of Medicine
| | | | - Kristen A Batich
- School of Medicine.,Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | - Adam M Swartz
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | | | | | | | - Karolina Woroniecka
- School of Medicine.,Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | - Smita K Nair
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and.,Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Peter E Fecci
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and
| | - John H Sampson
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and
| | - Michael D Gunn
- Department of Immunology.,Division of Cardiology, Department of Medicine
| |
Collapse
|
39
|
Abdouni Y, Yilmaz G, Monaco A, Aksakal R, Becer CR. Effect of Arm Number and Length of Star-Shaped Glycopolymers on Binding to Dendritic and Langerhans Cell Lectins. Biomacromolecules 2020; 21:3756-3764. [DOI: 10.1021/acs.biomac.0c00856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yamin Abdouni
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, United Kingdom
| | - Gokhan Yilmaz
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Alessandra Monaco
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, United Kingdom
| | - Resat Aksakal
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, United Kingdom
| | - C. Remzi Becer
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, United Kingdom
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| |
Collapse
|
40
|
Harnessing the Complete Repertoire of Conventional Dendritic Cell Functions for Cancer Immunotherapy. Pharmaceutics 2020; 12:pharmaceutics12070663. [PMID: 32674488 PMCID: PMC7408110 DOI: 10.3390/pharmaceutics12070663] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023] Open
Abstract
The onset of checkpoint inhibition revolutionized the treatment of cancer. However, studies from the last decade suggested that the sole enhancement of T cell functionality might not suffice to fight malignancies in all individuals. Dendritic cells (DCs) are not only part of the innate immune system, but also generals of adaptive immunity and they orchestrate the de novo induction of tolerogenic and immunogenic T cell responses. Thus, combinatorial approaches addressing DCs and T cells in parallel represent an attractive strategy to achieve higher response rates across patients. However, this requires profound knowledge about the dynamic interplay of DCs, T cells, other immune and tumor cells. Here, we summarize the DC subsets present in mice and men and highlight conserved and divergent characteristics between different subsets and species. Thereby, we supply a resource of the molecular players involved in key functional features of DCs ranging from their sentinel function, the translation of the sensed environment at the DC:T cell interface to the resulting specialized T cell effector modules, as well as the influence of the tumor microenvironment on the DC function. As of today, mostly monocyte derived dendritic cells (moDCs) are used in autologous cell therapies after tumor antigen loading. While showing encouraging results in a fraction of patients, the overall clinical response rate is still not optimal. By disentangling the general aspects of DC biology, we provide rationales for the design of next generation DC vaccines enabling to exploit and manipulate the described pathways for the purpose of cancer immunotherapy in vivo. Finally, we discuss how DC-based vaccines might synergize with checkpoint inhibition in the treatment of malignant diseases.
Collapse
|
41
|
Baldin AV, Savvateeva LV, Bazhin AV, Zamyatnin AA. Dendritic Cells in Anticancer Vaccination: Rationale for Ex Vivo Loading or In Vivo Targeting. Cancers (Basel) 2020; 12:cancers12030590. [PMID: 32150821 PMCID: PMC7139354 DOI: 10.3390/cancers12030590] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) have shown great potential as a component or target in the landscape of cancer immunotherapy. Different in vivo and ex vivo strategies of DC vaccine generation with different outcomes have been proposed. Numerous clinical trials have demonstrated their efficacy and safety in cancer patients. However, there is no consensus regarding which DC-based vaccine generation method is preferable. A problem of result comparison between trials in which different DC-loading or -targeting approaches have been applied remains. The employment of different DC generation and maturation methods, antigens and administration routes from trial to trial also limits the objective comparison of DC vaccines. In the present review, we discuss different methods of DC vaccine generation. We conclude that standardized trial designs, treatment settings and outcome assessment criteria will help to determine which DC vaccine generation approach should be applied in certain cancer cases. This will result in a reduction in alternatives in the selection of preferable DC-based vaccine tactics in patient. Moreover, it has become clear that the application of a DC vaccine alone is not sufficient and combination immunotherapy with recent advances, such as immune checkpoint inhibitors, should be employed to achieve a better clinical response and outcome.
Collapse
Affiliation(s)
- Alexey V. Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Lyudmila V. Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University of Munich, 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Department of Cell Signaling, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +74-956-229-843
| |
Collapse
|
42
|
Corgnac S, Botelho NK, Donda A, Romero P. Recombinant fusion proteins for targeting dendritic cell subsets in therapeutic cancer vaccine. Methods Enzymol 2020; 632:521-543. [DOI: 10.1016/bs.mie.2019.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
43
|
Pires CF, Rosa FF, Kurochkin I, Pereira CF. Understanding and Modulating Immunity With Cell Reprogramming. Front Immunol 2019; 10:2809. [PMID: 31921109 PMCID: PMC6917620 DOI: 10.3389/fimmu.2019.02809] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cell reprogramming concepts have been classically developed in the fields of developmental and stem cell biology and are currently being explored for regenerative medicine, given its potential to generate desired cell types for replacement therapy. Cell fate can be experimentally reversed or modified by enforced expression of lineage specific transcription factors leading to pluripotency or attainment of another somatic cell type identity. The possibility to reprogram fibroblasts into induced dendritic cells (DC) competent for antigen presentation creates a paradigm shift for understanding and modulating the immune system with direct cell reprogramming. PU.1, IRF8, and BATF3 were identified as sufficient and necessary to impose DC fate in unrelated cell types, taking advantage of Clec9a, a C-type lectin receptor with restricted expression in conventional DC type 1. The identification of such minimal gene regulatory networks helps to elucidate the molecular mechanisms governing development and lineage heterogeneity along the hematopoietic hierarchy. Furthermore, the generation of patient-tailored reprogrammed immune cells provides new and exciting tools for the expanding field of cancer immunotherapy. Here, we summarize cell reprogramming concepts and experimental approaches, review current knowledge at the intersection of cell reprogramming with hematopoiesis, and propose how cell fate engineering can be merged to immunology, opening new opportunities to understand the immune system in health and disease.
Collapse
Affiliation(s)
- Cristiana F. Pires
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Fábio F. Rosa
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ilia Kurochkin
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Carlos-Filipe Pereira
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
44
|
Lu Y, Xu W, Gu Y, Chang X, Wei G, Rong Z, Qin L, Chen X, Zhou F. Non-small Cell Lung Cancer Cells Modulate the Development of Human CD1c + Conventional Dendritic Cell Subsets Mediated by CD103 and CD205. Front Immunol 2019; 10:2829. [PMID: 31921114 PMCID: PMC6914740 DOI: 10.3389/fimmu.2019.02829] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced non-small cell lung cancer (NSCLC) leads to a high death rate in patients and is a major threat to human health. NSCLC induces an immune suppressive microenvironment and escapes from immune surveillance in vivo. At present, the molecular mechanisms of NSCLC immunopathogenesis and the immune suppressive microenvironment induced by NSCLC have not been fully elucidated. Here, we focus on the effect of NSCLC cells on the development and differentiation of human CD1c+ conventional dendritic cell (DC) subsets mediated by CD205 and CD103. The peripheral blood mononuclear cells (PBMCs) were isolated from NSCLC patients and healthy donors. DCs were induced and cocultured with primary NSCLC cells or tumor cell line H1299. DCs without incubation with tumor cells are control. The protein expression of costimulatory molecules such as CD80 and CD86, HLA-DR, pro-/anti-inflammatory cytokines such as IL-10 and IL-12, and CD205 and CD103 on CD1c+ DCs was detected by flow cytometry. Our data revealed two new subpopulations of human CD1c+ DCs (CD1c+CD205+CD103+ and CD1c+CD205+CD103− DC) in healthy donors and NSCLC patients. NSCLC cells modulate the development of the CD1c+CD205+CD103+ DC and CD1c+CD205+CD103− DC subpopulations in vitro and ex vivo. NSCLC cells also suppress the expression of signal molecules such as CD40, CD80, CD86, and HLA-DR on CD1c+ DCs. In addition, the production of pro-inflammatory cytokines, including IL-12 and IL-23, is downregulated by NSCLC cells; however, the secretion of anti-inflammatory cytokines, such as IL-10 and IL-27, by CD1c+ DCs is upregulated by NSCLC cells. Our results suggest that NSCLC cells may induce immune tolerogenic DCs, which block DC-mediated anti-tumor immunity in NSCLC patients. Our data may be helpful in revealing new cellular mechanisms related to the induction of tolerogenic CD1c+ DCs by NSCLCs and the development of an immune suppressive microenvironment that causes tumor cells to escape immune surveillance. Our results indicate a potential role for CD1c+ DC subsets mediated by CD205 and CD103 in DC-mediated immunotherapy to target NSCLC in the future.
Collapse
Affiliation(s)
- Yong Lu
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Wenlong Xu
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Yanli Gu
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Xu Chang
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Guojian Wei
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Zhien Rong
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Li Qin
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| | - Xiaoping Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China.,Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Fang Zhou
- Department of Experimental and Clinical Immunology, CAS Lamvac Biotech Co., Ltd., Guangzhou, China
| |
Collapse
|
45
|
Merlino G, Fiascarelli A, Bigioni M, Bressan A, Carrisi C, Bellarosa D, Salerno M, Bugianesi R, Manno R, Bernadó Morales C, Arribas J, Dusek RL, Ackroyd JE, Pham PH, Awdew R, Aud D, Trang M, Lynch CM, Terrett J, Wilson KE, Rohlff C, Manzini S, Pellacani A, Binaschi M. MEN1309/OBT076, a First-In-Class Antibody-Drug Conjugate Targeting CD205 in Solid Tumors. Mol Cancer Ther 2019; 18:1533-1543. [PMID: 31227646 DOI: 10.1158/1535-7163.mct-18-0624] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 01/16/2019] [Accepted: 06/17/2019] [Indexed: 11/16/2022]
Abstract
CD205 is a type I transmembrane glycoprotein and is a member of the C-type lectin receptor family. Analysis by mass spectrometry revealed that CD205 was robustly expressed and highly prevalent in a variety of solid malignancies from different histotypes. IHC confirmed the increased expression of CD205 in pancreatic, bladder, and triple-negative breast cancer (TNBC) compared with that in the corresponding normal tissues. Using immunofluorescence microscopy, rapid internalization of the CD205 antigen was observed. These results supported the development of MEN1309/OBT076, a fully humanized CD205-targeting mAb conjugated to DM4, a potent maytansinoid derivate, via a cleavable N-succinimidyl-4-(2-pyridyldithio) butanoate linker. MEN1309/OBT076 was characterized in vitro for target binding affinity, mechanism of action, and cytotoxic activity against a panel of cancer cell lines. MEN1309/OBT076 displayed selective and potent cytotoxic effects against tumor cells exhibiting strong and low to moderate CD205 expression. In vivo, MEN1309/OBT076 showed potent antitumor activity resulting in durable responses and complete tumor regressions in many TNBC, pancreatic, and bladder cancer cell line-derived and patient-derived xenograft models, independent of antigen expression levels. Finally, the pharmacokinetics and pharmacodynamic profile of MEN1309/OBT076 was characterized in pancreatic tumor-bearing mice, demonstrating that the serum level of antibody-drug conjugate (ADC) achieved through dosing was consistent with the kinetics of its antitumor activity. Overall, our data demonstrate that MEN1309/OBT076 is a novel and selective ADC with potent activity against CD205-positive tumors. These data supported the clinical development of MEN1309/OBT076, and further evaluation of this ADC is currently ongoing in the first-in-human SHUTTLE clinical trial.
Collapse
Affiliation(s)
- Giuseppe Merlino
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy.
| | - Alessio Fiascarelli
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy
| | - Mario Bigioni
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy
| | - Alessandro Bressan
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy
| | - Corrado Carrisi
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy
| | - Daniela Bellarosa
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy
| | - Massimiliano Salerno
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy
| | - Rossana Bugianesi
- Department of Pharmacokinetics and Metabolism, Menarini Ricerche, Pomezia, Rome, Italy
| | | | - Cristina Bernadó Morales
- Preclinical Research Program, Vall D'Hebron, Institute of Oncology and Centro de Investigación Biomédica en Red en Oncologia (CIBERONC), Barcelona, Spain
| | - Joaquin Arribas
- Preclinical Research Program, Vall D'Hebron, Institute of Oncology and Centro de Investigación Biomédica en Red en Oncologia (CIBERONC), Barcelona, Spain.,Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, Campus de la UAB, Bellaterra, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | | | | | | | - Rahel Awdew
- Oxford BioTherapeutics, Ltd., Abingdon, United Kingdom
| | - Dee Aud
- Oxford BioTherapeutics, Ltd., Abingdon, United Kingdom
| | - Michael Trang
- Oxford BioTherapeutics, Ltd., Abingdon, United Kingdom
| | | | | | | | | | | | | | - Monica Binaschi
- Department of Experimental and Translational Oncology, Menarini Ricerche SpA, Pomezia, Rome, Italy
| |
Collapse
|
46
|
Targeting M2e to DEC-205 induces an enhanced serum antibody-dependent heterosubtypic protection against influenza A virus infection. Vaccine 2019; 37:2624-2633. [DOI: 10.1016/j.vaccine.2019.02.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/23/2019] [Accepted: 02/15/2019] [Indexed: 01/07/2023]
|
47
|
CD205-positive, Sepharose-induced peritoneal exudate cells: a new resource for DC research in the chicken. Vet Res Commun 2019; 43:115-122. [PMID: 30989431 DOI: 10.1007/s11259-019-09751-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/22/2019] [Indexed: 10/27/2022]
Abstract
Dendritic cells (DC) are important antigen-presenting cells and are among the least characterized immune cells in the chicken. In order to obtain chicken DC, current protocols require isolation of bone marrow myeloid progenitor cells and induction of DC differentiation with supplemental cytokines or negative selection of splenic cell preparations. Chicken peritoneal exudate cells (PEC) have traditionally been a source of various immune cells for ex vivo studies, primarily to investigate heterophils and macrophages. In this study, we observe the presence of CD205+ PEC populations, a marker of DC, as an additional resource to isolate and study chicken primary DCs. A panel of monoclonal antibodies was developed against the chicken CD205 DC marker and used to isolate CD205+ DC from the PEC population using magnetic bead cell sorting. This study reports the development of new anti-CD205 monoclonal antibodies as a reagent for chicken DC research, as well as PEC as a potential source of CD205+ DC for ex vivo studies in the chicken.
Collapse
|
48
|
Use of Dendritic Cell Receptors as Targets for Enhancing Anti-Cancer Immune Responses. Cancers (Basel) 2019; 11:cancers11030418. [PMID: 30909630 PMCID: PMC6469018 DOI: 10.3390/cancers11030418] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
A successful anti-cancer vaccine construct depends on its ability to induce humoral and cellular immunity against a specific antigen. Targeting receptors of dendritic cells to promote the loading of cancer antigen through an antibody-mediated antigen uptake mechanism is a promising strategy in cancer immunotherapy. Researchers have been targeting different dendritic cell receptors such as Fc receptors (FcR), various C-type lectin-like receptors such as dendritic and thymic epithelial cell-205 (DEC-205), dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), and Dectin-1 to enhance the uptake process and subsequent presentation of antigen to T cells through major histocompatibility complex (MHC) molecules. In this review, we compare different subtypes of dendritic cells, current knowledge on some important receptors of dendritic cells, and recent articles on targeting those receptors for anti-cancer immune responses in mouse models.
Collapse
|
49
|
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
|
50
|
Botelho NK, Tschumi BO, Hubbell JA, Swartz MA, Donda A, Romero P. Combination of Synthetic Long Peptides and XCL1 Fusion Proteins Results in Superior Tumor Control. Front Immunol 2019; 10:294. [PMID: 30863405 PMCID: PMC6399421 DOI: 10.3389/fimmu.2019.00294] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 02/05/2019] [Indexed: 12/24/2022] Open
Abstract
Cross-presenting Xcr1+CD8α DCs are attractive APCs to target for therapeutic cancer vaccines, as they are able to take up and process antigen from dying tumor cells for their MHCI-restricted presentation to CD8 T cells. To this aim, we developed fusion proteins made of the Xcr1 ligand Xcl1 fused to an OVA synthetic long peptide (SLP) and IgG1 Fc fragment. We demonstrated the specific binding and uptake of the Xcl1 fusion proteins by Xcr1+ DCs. Most importantly, their potent adjuvant effect on the H-2Kb/OVA specific T cell response was associated with a sustained tumor control even against the poorly immunogenic B16-OVA melanoma tumor. The increased tumor protection correlated with higher tumor infiltration of antigen-specific CD8+ T cells, increased IFNγ production and degranulation potential. Altogether, these results demonstrate that therapeutic cancer vaccines may be greatly improved by the combination of SLP antigen and Xcl1 fusion proteins.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CHO Cells
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Chemokines, C/genetics
- Chemokines, C/immunology
- Chemokines, C/metabolism
- Cricetinae
- Cricetulus
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/therapy
- Mice, Inbred C57BL
- Mice, Knockout
- Ovalbumin/genetics
- Ovalbumin/immunology
- Ovalbumin/metabolism
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Vaccination/methods
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Natalia K. Botelho
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Benjamin O. Tschumi
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Jeffrey A. Hubbell
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Melody A. Swartz
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, United States
| | - Alena Donda
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Pedro Romero
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|