Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (49)

Search Parameters:
Keywords = immunopeptidomics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 8964 KB  
Article
Immunopeptidome Landscape During Brucella melitensis Infection in Mice
by Jing Jin, Yaming Sheng, Tingting Li, Kang Wang, Fanghao Geng, Yi Li and Jianfeng Gao
Int. J. Mol. Sci. 2025, 26(18), 8874; https://doi.org/10.3390/ijms26188874 - 12 Sep 2025
Viewed by 489
Abstract
Mouse bone marrow-derived dendritic cells (BMDCs) were infected in vitro with the recombinant Brucella melitensis M5+gfp strain. The immunopeptidome of Brucella melitensis M5+gfp, which presented peptides bound to MHC class II molecules on their surface, was isolated and characterized. BMDCs [...] Read more.
Mouse bone marrow-derived dendritic cells (BMDCs) were infected in vitro with the recombinant Brucella melitensis M5+gfp strain. The immunopeptidome of Brucella melitensis M5+gfp, which presented peptides bound to MHC class II molecules on their surface, was isolated and characterized. BMDCs infected with Brucella melitensis M5+gfp were subjected to hypotonic lysis. The associated immunopeptidome was then isolated and characterized using co-immunoprecipitation (Co-IP) coupled with liquid chromatography–tandem mass spectrometry (LC-MS/MS). A total of 289 MHC-II-bound Brucella melitensis M5+gfp peptide sequences were identified, mapping to 183 distinct proteins. We successfully define the Brucella melitensis M5+gfp immunopeptidome presented by MHC-II on infected BMDCs. The source proteins of these peptides exhibited significant abundance and functional, structural, and pathway diversity. This study demonstrates that during antigen presentation by antigen-presenting cells (APCs), Brucella melitensis peptides presented by MHC-II originate from a broad repertoire of proteins, not limited to surface antigens. This complex immunopeptidome, shaped by active selection mechanisms, provides diverse targets for host immune recognition. These findings establish a foundation for further investigation into the transfer of comprehensive immune information between immune cells and the elicitation of immune responses. This work also paves the way for identifying specific T-cell receptors involved in recognition and immune activation, thereby facilitating the analysis of adaptive immunity’s molecular basis. Furthermore, this study provides an innovative approach for immunopeptidome analysis, providing a crucial theoretical foundation for developing novel Brucella melitensis subunit vaccines. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

26 pages, 635 KB  
Review
Decoding Immunodeficiencies with Artificial Intelligence: A New Era of Precision Medicine
by Raffaele Sciaccotta, Paola Barone, Giuseppe Murdaca, Manlio Fazio, Fabio Stagno, Sebastiano Gangemi, Sara Genovese and Alessandro Allegra
Biomedicines 2025, 13(8), 1836; https://doi.org/10.3390/biomedicines13081836 - 28 Jul 2025
Viewed by 889
Abstract
Primary and secondary immunodeficiencies comprise a wide array of illnesses marked by immune system abnormalities, resulting in heightened vulnerability to infections, autoimmunity, and cancers. Notwithstanding progress in diagnostic instruments and an enhanced comprehension of the underlying pathophysiology, delayed diagnosis and underreporting persist as [...] Read more.
Primary and secondary immunodeficiencies comprise a wide array of illnesses marked by immune system abnormalities, resulting in heightened vulnerability to infections, autoimmunity, and cancers. Notwithstanding progress in diagnostic instruments and an enhanced comprehension of the underlying pathophysiology, delayed diagnosis and underreporting persist as considerable obstacles. The implementation of artificial intelligence into clinical practice has surfaced as a viable method to enhance early detection, risk assessment, and management of immunodeficiencies. Recent advancements illustrate how artificial intelligence-driven models, such as predictive algorithms, electronic phenotyping, and automated flow cytometry analysis, might enable early diagnosis, minimize diagnostic delays, and enhance personalized treatment methods. Furthermore, artificial intelligence-driven immunopeptidomics and phenotypic categorization are enhancing vaccine development and biomarker identification. Successful implementation necessitates overcoming problems associated with data standardization, model validation, and ethical issues. Future advancements will necessitate a multidisciplinary partnership among physicians, data scientists, and governments to effectively use the revolutionary capabilities of artificial intelligence, therefore ushering in an age of precision medicine in immunodeficiencies. Full article
(This article belongs to the Section Immunology and Immunotherapy)
Show Figures

Figure 1

19 pages, 1427 KB  
Article
Citrullinated ENO1 Vaccine Enhances PD-1 Blockade in Mice Implanted with Murine Triple-Negative Breast Cancer Cells
by Ricardo A. León-Letelier, Alejandro M. Sevillano-Mantas, Yihui Chen, Soyoung Park, Jody Vykoukal, Johannes F. Fahrmann, Edwin J. Ostrin, Candace Garrett, Rongzhang Dou, Yining Cai, Fu-Chung Hsiao, Jennifer B. Dennison, Eduardo Vilar, Banu K. Arun, Samir Hanash and Hiroyuki Katayama
Vaccines 2025, 13(6), 629; https://doi.org/10.3390/vaccines13060629 - 11 Jun 2025
Viewed by 1521
Abstract
Background/Objectives:Cancer vaccine targets mostly include mutations and overexpressed proteins. However, cancer-associated post-translational modifications (PTMs) may also induce immune responses. Previously, our group established the enzyme protein arginine deiminase type-2 (PADI2), which catalyzes citrullination modification, is highly expressed in triple-negative breast cancer (TNBC), [...] Read more.
Background/Objectives:Cancer vaccine targets mostly include mutations and overexpressed proteins. However, cancer-associated post-translational modifications (PTMs) may also induce immune responses. Previously, our group established the enzyme protein arginine deiminase type-2 (PADI2), which catalyzes citrullination modification, is highly expressed in triple-negative breast cancer (TNBC), promoting antigenicity. Methods: Here, we show the workflow of designing citrullinated enolase 1 (citENO1) vaccine peptides identified from breast cancer cells by mass spectrometry and demonstrate TNBC vaccine efficacy in the mouse model. Immunized mice with citENO1 peptides or the corresponding unmodified peptides, plus Poly I:C as an adjuvant, were orthotopically implanted with a TNBC murine cell line. Results: Vaccination with citENO1, but not unmodified ENO1 (umENO1), induced a greater percentage of activated CD8+ PD-1+ T cells and effector memory T cells in skin-draining lymph nodes (SDLNs). Remarkably, the citENO1 vaccine delayed tumor growth and prolonged overall survival, which was further enhanced by PD-1 blockade. Conclusions: Our data suggest that cancer-restricted post-translational modifications provide a source of vaccines that induce an anti-cancer immune response. Full article
(This article belongs to the Special Issue Personalised Cancer Vaccines)
Show Figures

Figure 1

14 pages, 3293 KB  
Article
The Validation of Antibodies Suitable for Flow Cytometric Analysis and Immunopeptidomics of Peptide–MHC Complexes in the Outbred Swiss Albino Mouse Strain
by Shanzou Chung, Isambard G. Knox-Johnson, Sarah E. Gazzard, Runqiu Song, Ngoc H. Le, Luise A. Cullen-McEwen, John F. Bertram, Anthony W. Purcell and Asolina Braun
Methods Protoc. 2025, 8(3), 43; https://doi.org/10.3390/mps8030043 - 24 Apr 2025
Viewed by 1331
Abstract
Antigen presentation on major histocompatibility complex (MHC) molecules is central to the initiation of immune responses, and a lot of our understanding about the antigen processing and presentation pathway has been gained through studies in mice. MHC molecules are the most genetically diverse [...] Read more.
Antigen presentation on major histocompatibility complex (MHC) molecules is central to the initiation of immune responses, and a lot of our understanding about the antigen processing and presentation pathway has been gained through studies in mice. MHC molecules are the most genetically diverse genes; consequently, mouse strains differ substantially in their MHC make up and resulting antigen presentation. Swiss mice are commonly used in pharmacological research, yet our understanding of antigen presentation in this strain is surprisingly limited. Here, we have tested a range of anti-MHC antibodies and present a range of clones suitable to analyse MHC class I and class II molecules in Swiss mice who have the H2-q MHC haplotype. Moreover, we demonstrate using immunopeptidomics that clones 28-12-8, 34-1-2, MKD6, and N22 are also suited to isolate MHC class I and class II ligands in this mouse strain. Thus, this work also establishes a first experimental account of the H2-q-derived thymus and spleen immunopeptidome in Swiss mice which bears strong resemblance with ligands isolated from the H2-d MHC haplotype of Balb/C mice. The analysis of source proteins shows common but also organ- and function-specific antigen presentation in line with the involvement of the thymus in tolerance induction and the function of the spleen as a site of immune responses. Full article
(This article belongs to the Section Molecular and Cellular Biology)
Show Figures

Figure 1

28 pages, 6528 KB  
Article
Endoplasmic Reticulum-Targeted Phototherapy Remodels the Tumor Immunopeptidome to Enhance Immunogenic Cell Death and Adaptive Anti-Tumor Immunity
by Weidong Xiao, Mingquan Gao, Banghui Mo, Xie Huang, Zaizhi Du, Shufeng Wang, Jianhong Chen, Shenglin Luo and Haiyan Xing
Pharmaceuticals 2025, 18(4), 491; https://doi.org/10.3390/ph18040491 - 28 Mar 2025
Cited by 2 | Viewed by 1120
Abstract
Background: Endoplasmic reticulum (ER)-targeted phototherapy has emerged as a promising approach to amplify ER stress, induce immunogenic cell death (ICD), and enhance anti-tumor immunity. However, its impact on the antigenicity of dying tumor cells remains poorly understood. Methods: Laser activation of the ER-targeted [...] Read more.
Background: Endoplasmic reticulum (ER)-targeted phototherapy has emerged as a promising approach to amplify ER stress, induce immunogenic cell death (ICD), and enhance anti-tumor immunity. However, its impact on the antigenicity of dying tumor cells remains poorly understood. Methods: Laser activation of the ER-targeted photosensitizer ER-Cy-poNO2 was performed to investigate its effects on tumor cell antigenicity. Transcriptomic analysis was carried out to assess gene expression changes. Immunopeptidomics profiling was used to identify high-affinity major histocompatibility complex class I (MHC-I) ligands. In vitro functional studies were conducted to evaluate dendritic cell maturation and T lymphocyte activation, while in vivo experiments were performed by combining the identified peptide with poly IC to evaluate anti-tumor immunity. Results: Laser activation of ER-Cy-poNO2 significantly remodeled the antigenic landscape of 4T-1 tumor cells, enhancing their immunogenicity. Transcriptomic analysis revealed upregulation of antigen processing and presentation pathways. Immunopeptidomics profiling identified multiple high-affinity MHC-I ligands, with IF4G3986–994 (QGPKTIEQI) showing exceptional immunogenicity. In vitro, IF4G3986–994 promoted dendritic cell maturation and enhanced T lymphocytes activation. In vivo, the combination of IF4G3986–994 with poly IC elicited robust anti-tumor immunity, characterized by increased CD8+ T lymphocytes infiltration, reduced regulatory T cells (Tregs) in the tumor microenvironment, elevated systemic Interferon-gamma (IFN-γ) levels, and significant tumor growth inhibition without systemic toxicity. Conclusions: These findings establish a mechanistic link between ER stress-driven ICD, immunopeptidome remodeling, and adaptive immune activation, highlighting the potential of ER-targeted phototherapy as a platform for identifying immunogenic peptides and advancing peptide-based cancer vaccines. Full article
(This article belongs to the Special Issue Photodynamic Therapy: 3rd Edition)
Show Figures

Figure 1

15 pages, 1266 KB  
Review
Technology Innovation for Discovering Renal Autoantibodies in Autoimmune Conditions
by Maurizio Bruschi, Giovanni Candiano, Andrea Petretto, Andrea Angeletti, Pier Luigi Meroni, Marco Prunotto and Gian Marco Ghiggeri
Int. J. Mol. Sci. 2024, 25(23), 12659; https://doi.org/10.3390/ijms252312659 - 25 Nov 2024
Cited by 1 | Viewed by 1665
Abstract
Autoimmune glomerulonephritis is a homogeneous area of renal pathology with clinical relevance in terms of its numerical impact and difficulties in its treatment. Systemic lupus erythematosus/lupus nephritis and membranous nephropathy are the two most frequent autoimmune conditions with clinical relevance. They are characterized [...] Read more.
Autoimmune glomerulonephritis is a homogeneous area of renal pathology with clinical relevance in terms of its numerical impact and difficulties in its treatment. Systemic lupus erythematosus/lupus nephritis and membranous nephropathy are the two most frequent autoimmune conditions with clinical relevance. They are characterized by glomerular deposition of circulating autoantibodies that recognize glomerular antigens. Technologies for studying renal tissue and circulating antibodies have evolved over the years and have culminated with the direct analysis of antigen–antibody complexes in renal bioptic fragments. Initial studies utilized renal microdissection to obtain glomerular tissue. Obtaining immunoprecipitates after partial proteolysis of renal tissue is a recent evolution that eliminates the need for tissue microdissection. New technologies based on ‘super-resolution microscopy’ have added the possibility of a direct analysis of the interaction between circulating autoantibodies and their target antigens in glomeruli. Peptide and protein arrays represent the new frontier for identifying new autoantibodies in circulation. Peptide arrays consist of 7.5 million aligned peptides with 16 amino acids each, which cover the whole human proteome; protein arrays utilize, instead, a chip containing structured proteins, with 26.000 overall. An example of the application of the peptide array is the discovery in membranous nephropathy of many new circulating autoantibodies including formin-like-1, a protein of podosomes that is implicated in macrophage movements. Studies that utilize protein arrays are now in progress and will soon be published. The contribution of new technologies is expected to be relevant for extending our knowledge of the mechanisms involved in the pathogenesis of several autoimmune conditions. They may also add significant tools in clinical settings and modify the therapeutic handling of conditions that are not considered to be autoimmune. Full article
Show Figures

Figure 1

22 pages, 3581 KB  
Article
Immunopeptidomics of Salmonella enterica Serovar Typhimurium-Infected Pig Macrophages Genotyped for Class II Molecules
by Carmen Celis-Giraldo, Carlos F. Suárez, William Agudelo, Nieves Ibarrola, Rosa Degano, Jaime Díaz, Raúl Manzano-Román and Manuel A. Patarroyo
Biology 2024, 13(10), 832; https://doi.org/10.3390/biology13100832 - 16 Oct 2024
Cited by 1 | Viewed by 2428
Abstract
Salmonellosis is a zoonotic infection that has a major impact on human health; consuming contaminated pork products is the main source of such infection. Vaccination responses to classic vaccines have been unsatisfactory; that is why peptide subunit-based vaccines represent an excellent alternative. Immunopeptidomics [...] Read more.
Salmonellosis is a zoonotic infection that has a major impact on human health; consuming contaminated pork products is the main source of such infection. Vaccination responses to classic vaccines have been unsatisfactory; that is why peptide subunit-based vaccines represent an excellent alternative. Immunopeptidomics was used in this study as a novel approach for identifying antigens coupled to major histocompatibility complex class II molecules. Three homozygous individuals having three different haplotypes (Lr-0.23, Lr-0.12, and Lr-0.21) were thus selected as donors; peripheral blood macrophages were then obtained and stimulated with Salmonella typhimurium (MOI 1:40). Although similarities were observed regarding peptide length distribution, elution patterns varied between individuals; in total, 1990 unique peptides were identified as follows: 372 for Pig 1 (Lr-0.23), 438 for Pig 2 (Lr.0.12) and 1180 for Pig 3 (Lr.0.21). Thirty-one S. typhimurium unique peptides were identified; most of the identified peptides belonged to outer membrane protein A and chaperonin GroEL. Notably, 87% of the identified bacterial peptides were predicted in silico to be elution ligands. These results encourage further in vivo studies to assess the immunogenicity of the identified peptides, as well as their usefulness as possible protective vaccine candidates. Full article
(This article belongs to the Section Infection Biology)
Show Figures

Graphical abstract

20 pages, 2746 KB  
Article
Identification of a Clade-Specific HLA-C*03:02 CTL Epitope GY9 Derived from the HIV-1 p17 Matrix Protein
by Samuel Kyobe, Savannah Mwesigwa, Gyaviira Nkurunungi, Gaone Retshabile, Moses Egesa, Eric Katagirya, Marion Amujal, Busisiwe C. Mlotshwa, Lesedi Williams, Hakim Sendagire, on behalf of the CAfGEN Consortium, Dithan Kiragga, Graeme Mardon, Mogomotsi Matshaba, Neil A. Hanchard, Jacqueline Kyosiimire-Lugemwa and David Robinson
Int. J. Mol. Sci. 2024, 25(17), 9683; https://doi.org/10.3390/ijms25179683 - 6 Sep 2024
Cited by 1 | Viewed by 1836
Abstract
Efforts towards an effective HIV-1 vaccine have remained mainly unsuccessful. There is increasing evidence for a potential role of HLA-C-restricted CD8+ T cell responses in HIV-1 control, including our recent report of HLA-C*03:02 among African children. However, there are no documented optimal [...] Read more.
Efforts towards an effective HIV-1 vaccine have remained mainly unsuccessful. There is increasing evidence for a potential role of HLA-C-restricted CD8+ T cell responses in HIV-1 control, including our recent report of HLA-C*03:02 among African children. However, there are no documented optimal HIV-1 CD8+ T cell epitopes restricted by HLA-C*03:02; additionally, the structural influence of HLA-C*03:02 on epitope binding is undetermined. Immunoinformatics approaches provide a fast and inexpensive method to discover HLA-restricted epitopes. Here, we employed immunopeptidomics to identify HLA-C*03:02 CD8+ T cell epitopes. We identified a clade-specific Gag-derived GY9 (GTEELRSLY) HIV-1 p17 matrix epitope potentially restricted to HLA-C*03:02. Residues E62, T142, and E151 in the HLA-C*03:02 binding groove and positions p3, p6, and p9 on the GY9 epitope are crucial in shaping and stabilizing the epitope binding. Our findings support the growing evidence of the contribution of HLA-C molecules to HIV-1 control and provide a prospect for vaccine strategies. Full article
Show Figures

Figure 1

22 pages, 3193 KB  
Article
Crucial Parameters for Immunopeptidome Characterization: A Systematic Evaluation
by Pablo Juanes-Velasco, Carlota Arias-Hidalgo, Marina L. García-Vaquero, Janet Sotolongo-Ravelo, Teresa Paíno, Quentin Lécrevisse, Alicia Landeira-Viñuela, Rafael Góngora, Ángela-Patricia Hernández and Manuel Fuentes
Int. J. Mol. Sci. 2024, 25(17), 9564; https://doi.org/10.3390/ijms25179564 - 3 Sep 2024
Cited by 1 | Viewed by 3099
Abstract
Immunopeptidomics is the area of knowledge focused on the study of peptides assembled in the major histocompatibility complex (MHC), or human leukocyte antigen (HLA) in humans, which could activate the immune response via specific and selective T cell recognition. Advances in high-sensitivity mass [...] Read more.
Immunopeptidomics is the area of knowledge focused on the study of peptides assembled in the major histocompatibility complex (MHC), or human leukocyte antigen (HLA) in humans, which could activate the immune response via specific and selective T cell recognition. Advances in high-sensitivity mass spectrometry have enabled the detailed identification and quantification of the immunopeptidome, significantly impacting fields like oncology, infections, and autoimmune diseases. Current immunopeptidomics approaches primarily focus on workflows to identify immunopeptides from HLA molecules, requiring the isolation of the HLA from relevant cells or tissues. Common critical steps in these workflows, such as cell lysis, HLA immunoenrichment, and peptide isolation, significantly influence outcomes. A systematic evaluation of these steps led to the creation of an ‘Immunopeptidome Score’ to enhance the reproducibility and robustness of these workflows. This score, derived from LC-MS/MS datasets (ProteomeXchange identifier PXD038165), in combination with available information from public databases, aids in optimizing the immunopeptidome characterization process. The ‘Immunopeptidome Score’ has been applied in a systematic analysis of protein extraction, HLA immunoprecipitation, and peptide recovery yields across several tumor cell lines enabling the selection of peptides with optimal features and, therefore, the identification of potential biomarker and therapeutic targets. Full article
(This article belongs to the Special Issue Advances in Proteomics in Cancer)
Show Figures

Figure 1

16 pages, 2168 KB  
Article
Identification of HLA-A*11:01 and A*02:01-Restricted EBV Peptides Using HLA Peptidomics
by Yufei Wang, Wanlin Zhang, Ruona Shi, Yanran Luo, Zhenhuan Feng, Yanhong Chen, Qiuting Zhang, Yan Zhou, Jingtong Liang, Xiaoping Ye, Qisheng Feng, Xiaofei Zhang and Miao Xu
Viruses 2024, 16(5), 669; https://doi.org/10.3390/v16050669 - 25 Apr 2024
Cited by 2 | Viewed by 4059
Abstract
Epstein-Barr Virus (EBV) is closely linked to nasopharyngeal carcinoma (NPC), notably prevalent in southern China. Although type II latency of EBV plays a crucial role in the development of NPC, some lytic genes and intermittent reactivation are also critical for viral propagation and [...] Read more.
Epstein-Barr Virus (EBV) is closely linked to nasopharyngeal carcinoma (NPC), notably prevalent in southern China. Although type II latency of EBV plays a crucial role in the development of NPC, some lytic genes and intermittent reactivation are also critical for viral propagation and tumor progression. Since T cell-mediated immunity is effective in targeted killing of EBV-positive cells, it is important to identify EBV-derived peptides presented by highly prevalent human leukocyte antigen class I (HLA-I) molecules throughout the EBV life cycle. Here, we constructed an EBV-positive NPC cell model to evaluate the presentation of EBV lytic phase peptides on streptavidin-tagged specific HLA-I molecules. Utilizing a mass spectrometry (LC-MS/MS)-based immunopeptidomic approach, we characterized eleven novel EBV peptides as well as two previously identified peptides. Furthermore, we determined these peptides were immunogenic and could stimulate PBMCs from EBV VCA/NA-IgA positive donors in an NPC endemic southern Chinese population. Overall, this work demonstrates that highly prevalent HLA-I-specific EBV peptides can be captured and functionally presented to elicit immune responses in an in vitro model, which provides insight into the epitopes presented during EBV lytic cycle and reactivation. It expands the range of viral targets for potential NPC early diagnosis and treatment. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

26 pages, 4298 KB  
Article
Birinapant Reshapes the Tumor Immunopeptidome and Enhances Antigen Presentation
by Weiyan Zhang, Shenghuan Sun, Wenyuan Zhu, Delan Meng, Weiyi Hu, Siqi Yang, Mingjie Gao, Pengju Yao, Yuhao Wang, Qingsong Wang and Jianguo Ji
Int. J. Mol. Sci. 2024, 25(7), 3660; https://doi.org/10.3390/ijms25073660 - 25 Mar 2024
Viewed by 2780
Abstract
Birinapant, an antagonist of the inhibitor of apoptosis proteins, upregulates MHCs in tumor cells and displays a better tumoricidal effect when used in combination with immune checkpoint inhibitors, indicating that Birinapant may affect the antigen presentation pathway; however, the mechanism remains elusive. Based [...] Read more.
Birinapant, an antagonist of the inhibitor of apoptosis proteins, upregulates MHCs in tumor cells and displays a better tumoricidal effect when used in combination with immune checkpoint inhibitors, indicating that Birinapant may affect the antigen presentation pathway; however, the mechanism remains elusive. Based on high-resolution mass spectrometry and in vitro and in vivo models, we adopted integrated genomics, proteomics, and immunopeptidomics strategies to study the mechanism underlying the regulation of tumor immunity by Birinapant from the perspective of antigen presentation. Firstly, in HT29 and MCF7 cells, Birinapant increased the number and abundance of immunopeptides and source proteins. Secondly, a greater number of cancer/testis antigen peptides with increased abundance and more neoantigens were identified following Birinapant treatment. Moreover, we demonstrate the existence and immunogenicity of a neoantigen derived from insertion/deletion mutation. Thirdly, in HT29 cell-derived xenograft models, Birinapant administration also reshaped the immunopeptidome, and the tumor exhibited better immunogenicity. These data suggest that Birinapant can reshape the tumor immunopeptidome with respect to quality and quantity, which improves the presentation of CTA peptides and neoantigens, thus enhancing the immunogenicity of tumor cells. Such changes may be vital to the effectiveness of combination therapy, which can be further transferred to the clinic or aid in the development of new immunotherapeutic strategies to improve the anti-tumor immune response. Full article
(This article belongs to the Special Issue Drug Discovery and Application of New Technologies)
Show Figures

Figure 1

19 pages, 1241 KB  
Review
Oncolytic Viruses in the Era of Omics, Computational Technologies, and Modeling: Thesis, Antithesis, and Synthesis
by Laura Menotti and Andrea Vannini
Int. J. Mol. Sci. 2023, 24(24), 17378; https://doi.org/10.3390/ijms242417378 - 12 Dec 2023
Viewed by 2671
Abstract
Oncolytic viruses (OVs) are the frontier therapy for refractory cancers, especially in integration with immunomodulation strategies. In cancer immunovirotherapy, the many available “omics” and systems biology technologies generate at a fast pace a challenging huge amount of data, where apparently clashing information mirrors [...] Read more.
Oncolytic viruses (OVs) are the frontier therapy for refractory cancers, especially in integration with immunomodulation strategies. In cancer immunovirotherapy, the many available “omics” and systems biology technologies generate at a fast pace a challenging huge amount of data, where apparently clashing information mirrors the complexity of individual clinical situations and OV used. In this review, we present and discuss how currently big data analysis, on one hand and, on the other, simulation, modeling, and computational technologies, provide invaluable support to interpret and integrate “omic” information and drive novel synthetic biology and personalized OV engineering approaches for effective immunovirotherapy. Altogether, these tools, possibly aided in the future by artificial intelligence as well, will allow for the blending of the information into OV recombinants able to achieve tumor clearance in a patient-tailored way. Various endeavors to the envisioned “synthesis” of turning OVs into personalized theranostic agents are presented. Full article
(This article belongs to the Special Issue Oncolytic Virotherapy 2.0)
Show Figures

Figure 1

11 pages, 675 KB  
Perspective
Immunopeptidomics in the Era of Single-Cell Proteomics
by Rupert L. Mayer and Karl Mechtler
Biology 2023, 12(12), 1514; https://doi.org/10.3390/biology12121514 - 12 Dec 2023
Cited by 9 | Viewed by 6625
Abstract
Immunopeptidomics, as the analysis of antigen peptides being presented to the immune system via major histocompatibility complexes (MHC), is being seen as an imperative tool for identifying epitopes for vaccine development to treat cancer and viral and bacterial infections as well as parasites. [...] Read more.
Immunopeptidomics, as the analysis of antigen peptides being presented to the immune system via major histocompatibility complexes (MHC), is being seen as an imperative tool for identifying epitopes for vaccine development to treat cancer and viral and bacterial infections as well as parasites. The field has made tremendous strides over the last 25 years but currently still faces challenges in sensitivity and throughput for widespread applications in personalized medicine and large vaccine development studies. Cutting-edge technological advancements in sample preparation, liquid chromatography as well as mass spectrometry, and data analysis, however, are currently transforming the field. This perspective showcases how the advent of single-cell proteomics has accelerated this transformation of immunopeptidomics in recent years and will pave the way for even more sensitive and higher-throughput immunopeptidomics analyses. Full article
(This article belongs to the Special Issue Proteomics in Immunology and Cell Signaling)
Show Figures

Figure 1

20 pages, 2725 KB  
Review
Targeting Proteasomes and the MHC Class I Antigen Presentation Machinery to Treat Cancer, Infections and Age-Related Diseases
by Priyanka S. Rana, James J. Ignatz-Hoover and James J. Driscoll
Cancers 2023, 15(23), 5632; https://doi.org/10.3390/cancers15235632 - 29 Nov 2023
Cited by 13 | Viewed by 6464
Abstract
The majority of T-cell responses involve proteasome-dependent protein degradation and the downstream presentation of oligopeptide products complexed with major histocompatibility complex (MHC) class I (MHC-I) molecules to peptide-restricted CD8+ T-cells. However, evasion of host immunity is a cancer hallmark that is achieved [...] Read more.
The majority of T-cell responses involve proteasome-dependent protein degradation and the downstream presentation of oligopeptide products complexed with major histocompatibility complex (MHC) class I (MHC-I) molecules to peptide-restricted CD8+ T-cells. However, evasion of host immunity is a cancer hallmark that is achieved by disruption of host antigen processing and presentation machinery (APM). Consequently, mechanisms of immune evasion promote cancer growth and survival as well as de novo and acquired resistance to immunotherapy. A multitude of cell signaling pathways modulate the APM and MHC-I-dependent antigen presentation. Pharmacologics that specifically target and modulate proteasome structure and activity represent a novel emerging strategy to improve the treatment of cancers and other diseases characterized by aberrant protein accumulation. FDA-approved pharmacologics that selectively activate proteasomes and/or immunoproteasomes can be repositioned to overcome the current bottlenecks that hinder drug development to enhance antigen presentation, modulate the immunopeptidome, and enhance the cytotoxic activity of endogenous or engineered T-cells. Strategies to enhance antigen presentation may also improve the antitumor activity of T-cell immunotherapies, checkpoint inhibitors, and cancer vaccines. Proteasomes represent actionable therapeutic targets to treat difficult-to-treat infectious processes and neurodegenerative diseases that are characterized by the unwanted accrual of insoluble, deleterious, and potentially toxic proteins. Taken together, we highlight the breadth and magnitude of the proteasome and the immense potential to amplify and unmask the immunopeptidomic landscape to improve the treatment of a spectrum of human diseases. Full article
(This article belongs to the Special Issue Inflammation, Immunity, and Cancer Progression)
Show Figures

Figure 1

25 pages, 6962 KB  
Article
Expanding the MAPPs Assay to Accommodate MHC-II Pan Receptors for Improved Predictability of Potential T Cell Epitopes
by Katharina Hartman, Guido Steiner, Michel Siegel, Cary M. Looney, Timothy P. Hickling, Katharine Bray-French, Sebastian Springer, Céline Marban-Doran and Axel Ducret
Biology 2023, 12(9), 1265; https://doi.org/10.3390/biology12091265 - 21 Sep 2023
Cited by 10 | Viewed by 3642
Abstract
A critical step in the immunogenicity cascade is attributed to human leukocyte antigen (HLA) II presentation triggering T cell immune responses. The liquid chromatography–tandem mass spectrometry (LC-MS/MS)-based major histocompatibility complex (MHC) II-associated peptide proteomics (MAPPs) assay is implemented during preclinical risk assessments to [...] Read more.
A critical step in the immunogenicity cascade is attributed to human leukocyte antigen (HLA) II presentation triggering T cell immune responses. The liquid chromatography–tandem mass spectrometry (LC-MS/MS)-based major histocompatibility complex (MHC) II-associated peptide proteomics (MAPPs) assay is implemented during preclinical risk assessments to identify biotherapeutic-derived T cell epitopes. Although studies indicate that HLA-DP and HLA-DQ alleles are linked to immunogenicity, most MAPPs studies are restricted to using HLA-DR as the dominant HLA II genotype due to the lack of well-characterized immunoprecipitating antibodies. Here, we address this issue by testing various commercially available clones of MHC-II pan (CR3/43, WR18, and Tü39), HLA-DP (B7/21), and HLA-DQ (SPV-L3 and 1a3) antibodies in the MAPPs assay, and characterizing identified peptides according to binding specificity. Our results reveal that HLA II receptor-precipitating reagents with similar reported specificities differ based on clonality and that MHC-II pan antibodies do not entirely exhibit pan-specific tendencies. Since no individual antibody clone is able to recover the complete HLA II peptide repertoire, we recommend a mixed strategy of clones L243, WR18, and SPV-L3 in a single immunoprecipitation step for more robust compound-specific peptide detection. Ultimately, our optimized MAPPs strategy improves the predictability and additional identification of T cell epitopes in immunogenicity risk assessments. Full article
(This article belongs to the Special Issue Proteomics in Immunology and Cell Signaling)
Show Figures

Figure 1

Back to TopTop