Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (59)

Search Parameters:
Keywords = alloreactive

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 7550 KB  
Article
Novel BCR-Targeting Fusion Proteins for Antigen-Specific Depletion of Alloreactive B Cells in Antibody-Mediated Rejection
by Jing Zhang, Leiyan Wei, Lei Song, Xiaofang Lu, Liang Tan, Xin Li, Li Fu, Qizhi Luo, Xubiao Xie and Yizhou Zou
Cells 2025, 14(18), 1410; https://doi.org/10.3390/cells14181410 - 9 Sep 2025
Viewed by 365
Abstract
Donor-specific anti-HLA antibodies (DSAs) bind to donor vascular endothelial cells and mediate allograft rejection (AMR), but a clinical challenge for which targeted therapeutic options remain limited. We used a multiplexed single-antigen bead (SAB) assay to detect anti-human leukocyte antigen (HLA) antibodies. Based on [...] Read more.
Donor-specific anti-HLA antibodies (DSAs) bind to donor vascular endothelial cells and mediate allograft rejection (AMR), but a clinical challenge for which targeted therapeutic options remain limited. We used a multiplexed single-antigen bead (SAB) assay to detect anti-human leukocyte antigen (HLA) antibodies. Based on the antigens which patient’s antibodies aganist to, we developed bivalent HLA-Fc fusion proteins composed of HLA-derived antigenic domains and human IgG1-Fc effector regions (rA24-Fc and rB13-Fc). Specific binding and functional activity of the HLA-Fc proteins were further validated by flow cytometry, ELISA, complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) assays. Our findings demonstrate that the fusion proteins rA24-Fc and rB13-Fc significantly reduced HLA-specific antibody reactivity in vitro. Notably, rA24-Fc and rB13-Fc selectively bound to B-cell hybridomas (e.g., mouse W6/32 cells) expressing membrane immunoglobulins (BCR) which bound to the most HLA class I antigens. Importantly, rA24-Fc and rB13-Fc elicited antigen-specific, Fc-dependent elimination of the specific B-cell hybridomas. This study highlights HLA-Fc fusion proteins as a promising therapeutic strategy for the antigen-specific suppression of depletion of alloreactive B cells through dual cytotoxic mechanisms. This precision targeted to BCR of B cells approach is used to apply to the treatment of antibody-mediated rejection. Full article
(This article belongs to the Special Issue Mechanisms of Immune Responses and Therapy)
Show Figures

Figure 1

15 pages, 1248 KB  
Article
In Vitro Silencing of MHC-I in Keratinocytes by Herpesvirus US11 Protein to Model Alloreactive Suppression
by Frederik Schlottmann, Sarah Strauß, Peter Maria Vogt and Vesna Bucan
Eur. Burn J. 2025, 6(3), 47; https://doi.org/10.3390/ebj6030047 - 21 Aug 2025
Viewed by 540
Abstract
Background: Secondary rejection remains a major obstacle in skin allografting. Some viruses, such as human herpesvirus and cytomegalovirus, evade immune detection through proteins like the unique short glycoprotein 11 (US11), which down-regulates major histocompatibility complex (MHC) class I expression. This study explores the [...] Read more.
Background: Secondary rejection remains a major obstacle in skin allografting. Some viruses, such as human herpesvirus and cytomegalovirus, evade immune detection through proteins like the unique short glycoprotein 11 (US11), which down-regulates major histocompatibility complex (MHC) class I expression. This study explores the use of recombinant US11 protein as a biopharmaceutical approach to reduce MHC-I expression and thus decrease alloreactivity in human primary keratinocytes. Methods: Human keratinocytes were treated with recombinant US11 protein, and MHC-I expression was assessed via Western blot and flow cytometry. To evaluate immunomodulatory effects, US11-stimulated keratinocytes were co-cultured with peripheral blood mononuclear cells (PBMCs), and interferon-gamma (IFN-γ) levels were measured by ELISA. Additionally, ex vivo human skin tissue was stimulated with US11 to assess long-term MHC-I modulation. Results: US11 treatment significantly reduced MHC-I surface expression in keratinocytes. Co-cultures showed decreased IFN-γ secretion, indicating lower T cell activation. Human skin tissue stimulated with US11 exhibited reduced MHC-I expression after 7 days. Conclusions: This proof-of-concept study suggests that recombinant US11 protein may serve as an effective biopharmaceutical to reduce keratinocyte immunogenicity. Further in vitro and in vivo studies are warranted to validate its potential for clinical application in skin transplantation. Full article
Show Figures

Figure 1

21 pages, 2365 KB  
Review
Natural Killer (NK) Cell Alloreactivity in Haploidentical Stem Cell Transplantation
by Mar Luis-Hidalgo, José Luis Piñana, Carlos Solano and Dolores Planelles
Cells 2025, 14(14), 1091; https://doi.org/10.3390/cells14141091 - 16 Jul 2025
Viewed by 671
Abstract
This paper conducts a literature review on the role of natural killer cells in haploidentical hematopoietic stem cell transplantation. Theoretical concepts related to KIR genes are introduced regarding their structure, nomenclature, genetic organization, polymorphism, and inheritance pattern, types of KIR proteins and receptors, [...] Read more.
This paper conducts a literature review on the role of natural killer cells in haploidentical hematopoietic stem cell transplantation. Theoretical concepts related to KIR genes are introduced regarding their structure, nomenclature, genetic organization, polymorphism, and inheritance pattern, types of KIR proteins and receptors, HLA ligands for KIR receptors, and the definition of different NK alloreactivity prediction models for the donor of haploidentical hematopoietic stem cell transplantation and the recipient. These models include the following and consider incompatibility: ligand–ligand, receptor–ligand, gene–gene, and KIR haplotype models or the KIR-B donor group. These models consider the presence or absence of specific ligands or receptors and/or KIR genes in the donor and recipient to predict alloreactivity. Determining the best model for predicting KIR alloreactivity and its significance in donor selection algorithms for haploidentical transplantation is still under investigation. Full article
(This article belongs to the Section Cellular Immunology)
Show Figures

Figure 1

18 pages, 2579 KB  
Article
Clinical Proof-of-Concept of a Non-Gene Editing Technology Using miRNA-Based shRNA to Engineer Allogeneic CAR T-Cells
by Caroline Lonez, Jennifer Bolsée, Fanny Huberty, Thuy Nguyen, Céline Jacques-Hespel, Sebastien Anguille, Anne Flament and Eytan Breman
Int. J. Mol. Sci. 2025, 26(4), 1658; https://doi.org/10.3390/ijms26041658 - 15 Feb 2025
Cited by 7 | Viewed by 2266
Abstract
With the success of chimeric antigen receptor (CAR) T-cell therapy in B-cell malignancies, efforts are being made to extend this therapy to other malignancies and broader patient populations. However, limitations associated with the time-consuming and highly personalized manufacturing of autologous CAR T-cells remain. [...] Read more.
With the success of chimeric antigen receptor (CAR) T-cell therapy in B-cell malignancies, efforts are being made to extend this therapy to other malignancies and broader patient populations. However, limitations associated with the time-consuming and highly personalized manufacturing of autologous CAR T-cells remain. Allogeneic CAR T-cell approaches may overcome these challenges but require further engineering to reduce their alloreactivity. As a means to prevent graft-versus-host disease (GvHD) of allogeneic CAR T-cells, we have selected a micro RNA (miRNA)-based short hairpin RNA (shRNA) targeting CD3ζ which efficiently downregulates the expression of the T-cell receptor (TCR) below detection level. We generated allogeneic anti-B-cell maturation antigen CAR T-cells (CYAD-211) that co-express an anti-CD3ζ miRNA-based shRNA within the CAR construct which efficiently inhibited TCR-mediated signaling in vitro and GvHD in vivo. CYAD-211 was subsequently evaluated in a Phase-I clinical trial (NCT04613557), in patients with relapsed or refractory multiple myeloma. No signs of GvHD were observed despite evidence of engraftment, demonstrating efficient downregulation of the TCR. Our data provide proof of concept that a non-gene-edited technology can generate fully functional allogeneic CAR T-cells, without any signs of GvHD. However, further engineering of the CAR T-cells is needed to improve their persistence and long-term activity. Full article
(This article belongs to the Special Issue Advanced Therapies for Inherited Diseases and Cancer: Recent Progress)
Show Figures

Graphical abstract

13 pages, 540 KB  
Article
Prospective Study of Recipient Human Leukocyte Antigen (HLA) Alloimmunization Following the Use of Cold-Stored Saphenous Vein Allografts in Vascular Surgery
by Elsa Madeleine Faure, Pascal Pedini, Caroline Bouchet, Pascal Branchereau, Catalin Cosma, Eric Picard and Christophe Picard
J. Clin. Med. 2025, 14(4), 1224; https://doi.org/10.3390/jcm14041224 - 13 Feb 2025
Viewed by 919
Abstract
Objectives: The aim of this study was to assess the HLA alloreactivity of cold-stored saphenous vein allografts (CSVAs) by identifying the production of HLA donor-specific antibodies (DSAs) in the recipient. The secondary objective was to evaluate CSVA rejection-related complications, such as CSVA thrombosis [...] Read more.
Objectives: The aim of this study was to assess the HLA alloreactivity of cold-stored saphenous vein allografts (CSVAs) by identifying the production of HLA donor-specific antibodies (DSAs) in the recipient. The secondary objective was to evaluate CSVA rejection-related complications, such as CSVA thrombosis and/or aneurysmal degeneration in the recipient. Methods: This was a single-center, prospective, experimental before-and-after study which included participants undergoing CSVA placement, either to create a vascular access (VA) for hemodialysis or to create a lower limb arterial bypass. On Day 1, before CSVA placement, total blood samples were taken for HLA typing by sequence-specific primers (SSPs) and anti-HLA antibody detection using a Luminex assay. One month after CSVA placement, a second blood sample was taken to assess the appearance of donor-specific antibodies or an increase in the level of anti-HLA antibodies. Patency of the CSVA and potential aneurysmal degeneration were evaluated at 3 and 6 months with a Doppler ultrasound checkup. Results: From September 2022 to November 2023, 45 patients were included (30 men, 67%; mean age: 71 ± 12 years). One month after CSVA placement, no appearance of de novo anti-HLA antibody was detected in anti-HLA antibody-negative patients at inclusion (n = 28). Among the patients who already had anti-HLA antibodies at inclusion (n = 17), no increase in anti-HLA antibody levels or appearance of de novo anti-HLA antibodies was detected. Conclusions: This prospective study evaluating the immunogenicity of CSVAs through the appearance of anti-HLA antibodies one month after placement demonstrates that they do not seem to induce any HLA alloreactivity. Therefore, they may be used without the risk of HLA immunization in patients awaiting organ transplantation. Full article
(This article belongs to the Section Immunology & Rheumatology)
Show Figures

Graphical abstract

21 pages, 7884 KB  
Article
SARS-CoV-2-Specific T-Cell as a Potent Therapeutic Strategy against Immune Evasion of Emerging COVID-19 Variants
by Keon-Il Im, Nayoun Kim, Junseok Lee, Ui-Hyeon Oh, Hye-Won Lee, Dong-Gun Lee, Gi-June Min, Raeseok Lee, Jinah Lee, Seungtaek Kim and Seok-Goo Cho
Int. J. Mol. Sci. 2024, 25(19), 10512; https://doi.org/10.3390/ijms251910512 - 29 Sep 2024
Cited by 2 | Viewed by 2936
Abstract
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing [...] Read more.
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing the protective abilities using virus-specific T cells (VSTs). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VSTs were generated by stimulating donor-derived peripheral blood mononuclear cells with spike, nucleocapsid, and membrane protein peptide mixtures. Phenotypic characterization, including T-cell receptor (TCR) vβ and pentamer analyses, was performed on the ex vivo-expanded cells. We infected human leukocyte antigen (HLA)-partially matched human Calu-3 cells with various authentic SARS-CoV-2 strains in a Biosafety Level 3 facility and co-cultured them with VSTs. VSTs exhibited a diverse TCR vβ repertoire, confirming their ability to target a broad range of SARS-CoV-2 antigens from both the ancestral and mutant strains, including Omicron BA.1 and BA.5. These ex vivo-expanded cells exhibited robust cytotoxicity and low alloreactivity against HLA-partially matched SARS-CoV-2-infected cells. Their cytotoxic effects were consistent across variants, targeting conserved spike and nucleocapsid epitopes. Our findings suggest that third-party partial HLA-matching VSTs could counter immune-escape mechanisms posed by emerging variants of concern. Full article
(This article belongs to the Special Issue Molecular Research and Insights into COVID-19: 2nd Edition)
Show Figures

Figure 1

12 pages, 2655 KB  
Article
Assessment of Corneal Graft Outcomes in a Murine Model of Endothelial Keratoplasty
by Akitomo Narimatsu, Rohan Bir Singh, Pier Luigi Surico, Seokjoo Lee, Katayoon Forouzanfar, Francesca Kahale, Aytan Musayeva, Thomas H. Dohlman, Tomas Blanco and Reza Dana
J. Clin. Med. 2024, 13(17), 5010; https://doi.org/10.3390/jcm13175010 - 24 Aug 2024
Viewed by 1991
Abstract
Objectives: In this study, we establish a protocol for evaluating the outcomes of endothelial keratoplasty, including graft survival, rejection, or failure. Additionally, we also evaluate the alloimmune response in graft recipients. Methods: We performed EK using C57BL/6 (allogeneic) and BALB/c (syngeneic) [...] Read more.
Objectives: In this study, we establish a protocol for evaluating the outcomes of endothelial keratoplasty, including graft survival, rejection, or failure. Additionally, we also evaluate the alloimmune response in graft recipients. Methods: We performed EK using C57BL/6 (allogeneic) and BALB/c (syngeneic) as donors and BALB/c mice as recipients. Slit-lamp examination and optical coherence tomography were performed for clinical evaluations for 16 weeks post-procedure. Criteria for the assessment of corneal opacity were established and the animals were graded weekly. Additionally, we assessed corneal endothelial cell density by harvesting the corneas and staining with zonula occludens-1 (ZO-1). Lastly, lymph nodes were collected, and CD4+ T cells were MACS-sorted and co-cultured with syngeneic or allogeneic antigen-presenting cells (APCs) to assess the IFN-γ expression levels by alloreactive Th1 cells (ELISPOT) in response to the direct (donor) or indirect (host) pathways of sensitization. Results: We observed graft failure in four animals, including irreversible corneal opacity, graft detachment, and anterior synechiae in the first four weeks. The remaining animals were graded between 0 and 5 as per the established criteria. The total and graft corneal thickness and endothelial cell density progressively worsened with a higher grade of corneal opacity. The direct allosensitization of Th1 cells was significantly higher in mice with a higher grade of corneal opacity. At 16 weeks follow-up, the grafts remained stable with low opacity scores in syngeneic EK recipients; however, the opacity scores were higher and variable in allogeneic EK recipients. Conclusions: These findings establish a standardized protocol to assess the graft outcomes in a murine model of EK. Furthermore, we delineate the underlying immunological pathway that contributes to the immune-mediated rejection of grafts in this model. Full article
(This article belongs to the Special Issue New Insights into Corneal Regeneration and Transplantation)
Show Figures

Figure 1

20 pages, 938 KB  
Review
The Immune Resistance Signature of Acute Myeloid Leukemia and Current Immunotherapy Strategies
by Daniel J. Chandra, Bernhard Alber and Jennifer N. Saultz
Cancers 2024, 16(15), 2615; https://doi.org/10.3390/cancers16152615 - 23 Jul 2024
Cited by 5 | Viewed by 4990
Abstract
Acute myeloid leukemia (AML) is a complex hematopoietic clonal disorder with limited curative options beyond stem cell transplantation. The success of transplant is intimately linked with the graft versus leukemia effect from the alloreactive donor immune cells including, T and NK cells. The [...] Read more.
Acute myeloid leukemia (AML) is a complex hematopoietic clonal disorder with limited curative options beyond stem cell transplantation. The success of transplant is intimately linked with the graft versus leukemia effect from the alloreactive donor immune cells including, T and NK cells. The immune system plays a dynamic role in leukemia survival and resistance. Despite our growing understanding of the immune microenvironment, responses to immune-based therapies differ greatly between patients. Herein, we review the biology of immune evasion mechanisms in AML, discuss the current landscape of immunotherapeutic strategies, and discuss the implications of therapeutic targets. This review focuses on T and NK cell-based therapy, including modified and non-modified NK cells, CAR-T and CAR-NK cells, antibodies, and checkpoint blockades. Understanding the complex interchange between immune tolerance and the emergence of tumor resistance will improve patient outcomes. Full article
(This article belongs to the Special Issue Hematologic Malignancies: Challenges from Diagnosis to Treatment)
Show Figures

Figure 1

14 pages, 1835 KB  
Protocol
An Adaptable Protocol to Generate a Murine Enteroid–Macrophage Co-Culture System
by Viktoria Hentschel, Deepalakshmi Govindarajan, Thomas Seufferlein and Milena Armacki
Int. J. Mol. Sci. 2024, 25(14), 7944; https://doi.org/10.3390/ijms25147944 - 20 Jul 2024
Cited by 2 | Viewed by 2805
Abstract
Impairment of the intestinal epithelial barrier is frequently seen as collateral damage in various local and systemic inflammatory conditions. The inflammatory process is characterized by reciprocal interactions between the host intestinal epithelium and mucosal innate immune cells, e.g., macrophages. This article provides step-by-step [...] Read more.
Impairment of the intestinal epithelial barrier is frequently seen as collateral damage in various local and systemic inflammatory conditions. The inflammatory process is characterized by reciprocal interactions between the host intestinal epithelium and mucosal innate immune cells, e.g., macrophages. This article provides step-by-step instructions on how to set up a murine enteroid–macrophage co-culture by culturing cellular elements in proximity separated by a porous membrane. Unlike previously published co-culture systems, we have combined enteroids grown from C57BL6j mice with syngeneic bone marrow-derived macrophages to preclude potential allo-reactions between immune cells and epithelium. Transformation of intestinal crypts into proliferative enteroids was achieved by cultivation in Wnt3a-Noggin-R-Spondin-conditioned medium supplemented with ROCK inhibitor Y-27632. The differentiated phenotype was promoted by the use of the Wnt3-deprived EGF-Noggin-R-Spondin medium. The resulting co-culture of primary cells can be employed as a basic model to better understand the reciprocal relationship between intestinal epithelium and macrophages. It can be used for in vitro modelling of mucosal inflammation, mimicked by stimulation of macrophages either while being in co-culture or before being introduced into co-culture, to simulate enterogenic sepsis or systemic conditions affecting the intestinal tract. Full article
(This article belongs to the Special Issue Recent Research in Stem Cells to Organoids)
Show Figures

Figure 1

27 pages, 1356 KB  
Review
Haploidentical HSCT in the Treatment of Pediatric Hematological Disorders
by Anna Marszołek, Maria Leśniak, Anna Sekunda, Aleksander Siwek, Zuzanna Skiba, Monika Lejman and Joanna Zawitkowska
Int. J. Mol. Sci. 2024, 25(12), 6380; https://doi.org/10.3390/ijms25126380 - 9 Jun 2024
Cited by 5 | Viewed by 3843
Abstract
Allogeneic hematopoietic stem cell transplantation has become a treatment option for otherwise non-curative conditions, both malignant and benign, affecting children and adults. Nevertheless, the latest research has been focusing extensively on transplantation from related and unrelated haploidentical donors, suitable for patients requiring emergent [...] Read more.
Allogeneic hematopoietic stem cell transplantation has become a treatment option for otherwise non-curative conditions, both malignant and benign, affecting children and adults. Nevertheless, the latest research has been focusing extensively on transplantation from related and unrelated haploidentical donors, suitable for patients requiring emergent hematopoietic stem cell transplantation (HSCT) in the absence of an HLA-matched donor. Haploidentical HSCT (haplo-HSCT) can be an effective treatment for non-malignant pediatric disorders, such as primary immunodeficiencies or hemoglobinopathies, by enabling a much quicker selection of the appropriate donor for virtually all patients, low incidence of graft-versus-host disease (GVHD), and transplant-related mortality (TRM). Moreover, the outcomes of haplo-HSCT among children with hematological malignancies have improved radically. The most demanding tasks for clinicians are minimizing T-cell-mediated alloreactivity as well as early GVHD prevention. As a result, several T-cell depletion approaches, such as ex vivo T-cell depletion (TCD), and T-cell replete approaches, such as a combination of anti-thymocyte globulin (ATG), post-transplantation cyclophosphamide (PTCy), cyclosporine/tacrolimus, mycophenolate mofetil, or methotrexate, have been taken up. As more research is needed to establish the most beneficial form of therapy, haplo-HSCT is currently considered an alternative donor strategy for pediatric and adult patients with complications like viral and bacterial infections, invasive fungal disease, and GVHD. Full article
(This article belongs to the Special Issue Hematological Malignancies: Molecular Mechanisms and Therapy)
Show Figures

Figure 1

13 pages, 1227 KB  
Review
Histocompatibility Testing: A Fundamental Aspect of Renal Transplant Workup
by Vikash Chandra Mishra, Dinesh Chandra and Vimarsh Raina
Transplantology 2024, 5(2), 85-97; https://doi.org/10.3390/transplantology5020009 - 15 May 2024
Cited by 1 | Viewed by 6331
Abstract
Histocompatibility testing is pivotal in any renal transplantation workup, aimed at enhancing prospective donor recipient compatibility and improving transplant outcomes. The evolution and advancement of histocompatibility testing, particularly HLA typing, have significantly improved its precision. This study outlines the historical progression from serologic [...] Read more.
Histocompatibility testing is pivotal in any renal transplantation workup, aimed at enhancing prospective donor recipient compatibility and improving transplant outcomes. The evolution and advancement of histocompatibility testing, particularly HLA typing, have significantly improved its precision. This study outlines the historical progression from serologic to DNA-based HLA typing, emphasizing the role of HLA proteins in immune response. Anti-HLA antibodies, targeting HLA proteins, pose challenges in renal transplantation. Monitoring and managing these antibodies are critical for renal transplant success. Complement-dependent cytotoxicity crossmatch and flow cytometry crossmatch are essential techniques for assessing donor–recipient compatibility. Panel-reactive antibody assesses antibodies against a panel of donor antigens, often HLA. Higher PRA levels (percentage) complicate donor matching, requiring specialized protocols. Virtual crossmatch evaluates recipient anti-HLA antibodies against potential donors through synthetic beads. This approach predicts crossmatch outcomes by comparing antibody profiles, offering a valuable tool for the risk assessment of renal transplantation. Despite advancements, a comprehensive understanding of alloreactive immune responses requires a combination of assays, emphasizing the importance of a multifaceted approach in histocompatibility testing. This is an attempt to compile the relevant information, providing a basis for comparison in a clear and foundational format for histocompatibility testing laboratories. Full article
Show Figures

Figure 1

17 pages, 1428 KB  
Review
Proteomic Analysis of Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Systematic Review
by Illayaraja Krishnan, Alvin Man Lung Chan, Jia Xian Law, Min Hwei Ng, Jaime Jacqueline Jayapalan and Yogeswaran Lokanathan
Int. J. Mol. Sci. 2024, 25(10), 5340; https://doi.org/10.3390/ijms25105340 - 14 May 2024
Cited by 4 | Viewed by 3281
Abstract
Numerous challenges remain within conventional cell-based therapy despite the growing trend of stem cells used to treat various life-debilitating diseases. These limitations include batch-to-batch heterogeneity, induced alloreactivity, cell survival and integration, poor scalability, and high cost of treatment, thus hindering successful translation from [...] Read more.
Numerous challenges remain within conventional cell-based therapy despite the growing trend of stem cells used to treat various life-debilitating diseases. These limitations include batch-to-batch heterogeneity, induced alloreactivity, cell survival and integration, poor scalability, and high cost of treatment, thus hindering successful translation from lab to bedside. However, recent pioneering technology has enabled the isolation and enrichment of small extracellular vesicles (EVs), canonically known as exosomes. EVs are described as a membrane-enclosed cargo of functional biomolecules not limited to lipids, nucleic acid, and proteins. Interestingly, studies have correlated the biological role of MSC-EVs to the paracrine activity of MSCs. This key evidence has led to rigorous studies on MSC-EVs as an acellular alternative. Using EVs as a therapy was proposed as a model leading to improvements through increased safety; enhanced bioavailability due to size and permeability; reduced heterogeneity by selective and quantifiable properties; and prolonged shelf-life via long-term freezing or lyophilization. Yet, the identity and potency of EVs are still relatively unknown due to various methods of preparation and to qualify the final product. This is reflected by the absence of regulatory strategies overseeing manufacturing, quality control, clinical implementation, and product registration. In this review, the authors review the various production processes and the proteomic profile of MSC-EVs. Full article
(This article belongs to the Special Issue Exosomes and Non-Coding RNA Research in Health and Disease)
Show Figures

Figure 1

16 pages, 5280 KB  
Article
Novel JAK Inhibitors to Reduce Graft-Versus-Host Disease after Allogeneic Hematopoietic Cell Transplantation in a Preclinical Mouse Model
by Sena Kim, Peter Ruminski, Megh Singh, Karl Staser, Kidist Ashami, Julie Ritchey, Sora Lim, John F. DiPersio and Jaebok Choi
Molecules 2024, 29(8), 1801; https://doi.org/10.3390/molecules29081801 - 16 Apr 2024
Cited by 4 | Viewed by 2167
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a highly effective, well-established treatment for patients with various hematologic malignancies and non-malignant diseases. The therapeutic benefits of allo-HCT are mediated by alloreactive T cells in donor grafts. However, there is a significant risk of graft-versus-host disease [...] Read more.
Allogeneic hematopoietic cell transplantation (allo-HCT) is a highly effective, well-established treatment for patients with various hematologic malignancies and non-malignant diseases. The therapeutic benefits of allo-HCT are mediated by alloreactive T cells in donor grafts. However, there is a significant risk of graft-versus-host disease (GvHD), in which the donor T cells recognize recipient cells as foreign and attack healthy organs in addition to malignancies. We previously demonstrated that targeting JAK1/JAK2, mediators of interferon-gamma receptor (IFNGR) and IL-6 receptor signaling, in donor T cells using baricitinib and ruxolitinib results in a significant reduction in GvHD after allo-HCT. Furthermore, we showed that balanced inhibition of JAK1/JAK2 while sparing JAK3 is important for the optimal prevention of GvHD. Thus, we have generated novel JAK1/JAK2 inhibitors, termed WU derivatives, by modifying baricitinib. Our results show that WU derivatives have the potential to mitigate GvHD by upregulating regulatory T cells and immune reconstitution while reducing the frequencies of antigen-presenting cells (APCs) and CD80 expression on these APCs in our preclinical mouse model of allo-HCT. In addition, WU derivatives effectively downregulated CXCR3 and T-bet in primary murine T cells. In summary, we have generated novel JAK inhibitors that could serve as alternatives to baricitinib or ruxolitinib. Full article
Show Figures

Figure 1

25 pages, 994 KB  
Review
Allogeneic CAR-T Therapy Technologies: Has the Promise Been Met?
by Caroline Lonez and Eytan Breman
Cells 2024, 13(2), 146; https://doi.org/10.3390/cells13020146 - 12 Jan 2024
Cited by 41 | Viewed by 13296
Abstract
This last decade, chimeric antigen receptor (CAR) T-cell therapy has become a real treatment option for patients with B-cell malignancies, while multiple efforts are being made to extend this therapy to other malignancies and broader patient populations. However, several limitations remain, including those [...] Read more.
This last decade, chimeric antigen receptor (CAR) T-cell therapy has become a real treatment option for patients with B-cell malignancies, while multiple efforts are being made to extend this therapy to other malignancies and broader patient populations. However, several limitations remain, including those associated with the time-consuming and highly personalized manufacturing of autologous CAR-Ts. Technologies to establish “off-the-shelf” allogeneic CAR-Ts with low alloreactivity are currently being developed, with a strong focus on gene-editing technologies. Although these technologies have many advantages, they have also strong limitations, including double-strand breaks in the DNA with multiple associated safety risks as well as the lack of modulation. As an alternative, non-gene-editing technologies provide an interesting approach to support the development of allogeneic CAR-Ts in the future, with possibilities of fine-tuning gene expression and easy development. Here, we will review the different ways allogeneic CAR-Ts can be manufactured and discuss which technologies are currently used. The biggest hurdles for successful therapy of allogeneic CAR-Ts will be summarized, and finally, an overview of the current clinical evidence for allogeneic CAR-Ts in comparison to its autologous counterpart will be given. Full article
(This article belongs to the Special Issue Advances in Allogeneic Cell Therapy)
Show Figures

Graphical abstract

10 pages, 1434 KB  
Communication
Blinatumomab Redirects Donor Lymphocytes against CD19+ Acute Lymphoblastic Leukemia without Relevant Bystander Alloreactivity after Haploidentical Hematopoietic Stem Cell Transplantation
by Antonella Mancusi, Francesco Zorutti, Loredana Ruggeri, Samanta Bonato, Sara Tricarico, Tiziana Zei, Roberta Iacucci Ostini, Valerio Viglione, Rebecca Sembenico, Sofia Sciabolacci, Valeria Cardinali, Massimo Fabrizio Martelli, Cristina Mecucci, Alessandra Carotti, Maria Paola Martelli, Andrea Velardi and Antonio Pierini
Int. J. Mol. Sci. 2023, 24(22), 16105; https://doi.org/10.3390/ijms242216105 - 9 Nov 2023
Cited by 1 | Viewed by 2114
Abstract
Blinatumomab alone or with donor leukocyte infusions (DLI) has been used after allogeneic hematopoietic stem cell transplantation (HSCT) as a salvage therapy in relapsing patients with CD19+ hematological malignancies. It was effective in a fraction of them, with low incidence of Graft-versus-Host [...] Read more.
Blinatumomab alone or with donor leukocyte infusions (DLI) has been used after allogeneic hematopoietic stem cell transplantation (HSCT) as a salvage therapy in relapsing patients with CD19+ hematological malignancies. It was effective in a fraction of them, with low incidence of Graft-versus-Host Disease (GvHD). Immunosuppressive drugs used as GvHD prophylaxis hinder T cell function and reduce the efficacy of the treatment. Because T cell-depleted haploidentical HSCT with donor regulatory and conventional T cells (Treg/Tcon haploidentical HSCT) does not require post-transplant immunosuppression, it is an ideal platform for the concomitant use of blinatumomab and DLI. However, the risk of GvHD is high because the donor is haploidentical. We treated two patients with CD19+ acute lymphoblastic leukemia (ALL) who had relapsed after Treg/Tcon haploidentical HSCT with blinatumomab and DLI. Despite the mismatch for one HLA haplotype, they did not develop GvHD and achieved complete remission with negative minimal residual disease. Consistently, we found that blinatumomab did not enhance T cell alloreactivity in vitro. Eventually, the two patients relapsed again because of their high disease risk. This study suggests that treatment with blinatumomab and DLI can be feasible to treat relapse after haploidentical transplantation, and its pre-emptive use should be considered to improve efficacy. Full article
(This article belongs to the Special Issue Pathophysiology to Novel Therapeutic Approaches for Leukemia)
Show Figures

Figure 1

Back to TopTop