Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,659)

Search Parameters:
Keywords = immunosuppressive (IS) therapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 7975 KB  
Article
Targeting Pan-Cancer Stemness: Core Regulatory lncRNAs as Novel Therapeutic Vulnerabilities
by Shengcheng Deng, Yufan Yang, Dapeng Gao, Jiajun Gao and Yuanyan Xiong
Int. J. Mol. Sci. 2025, 26(23), 11684; https://doi.org/10.3390/ijms262311684 - 2 Dec 2025
Abstract
Tumor stemness represents a key biological process that drives tumor progression and therapeutic resistance across various cancer types. To systematically elucidate the regulatory roles of long non-coding RNAs (lncRNAs) in this process, we integrated bulk transcriptomic data from The Cancer Genome Atlas (TCGA) [...] Read more.
Tumor stemness represents a key biological process that drives tumor progression and therapeutic resistance across various cancer types. To systematically elucidate the regulatory roles of long non-coding RNAs (lncRNAs) in this process, we integrated bulk transcriptomic data from The Cancer Genome Atlas (TCGA) with publicly available pan-cancer single-cell transcriptomic atlases. Using machine-learning-based stemness metrics, we successfully quantified stemness features and identified unique lncRNA gene sets for each cancer type at the bulk data level. The high-stemness subtype exhibited enhanced proliferation, an immunosuppressive microenvironment, and profound metabolic reprogramming. Based on these findings, we constructed a robust prognostic model with remarkable predictive performance across multiple cancer types. At the single-cell resolution, we reconstructed the dynamic trajectory of stemness evolution, uncovering distinctive metabolic and cell-communication patterns within cancer stem cells (CSCs). This multi-scale analysis consistently nominated a core set of regulatory lncRNAs, including NEAT1 and MALAT1. Our work not only nominates potential targets for stemness-directed therapy but also provides a comprehensive framework for understanding lncRNA-driven mechanisms of cancer aggressiveness and resistance. Full article
(This article belongs to the Section Molecular Informatics)
28 pages, 1673 KB  
Review
Immunological Crossroads: Optimizing Antirejection Regimens to Sustain Antitumor Immunity in Liver Transplant Recipients with Hepatocellular Carcinoma
by Chao Zhang, Xin Yuan and Kunlin Xie
Cancers 2025, 17(23), 3871; https://doi.org/10.3390/cancers17233871 (registering DOI) - 2 Dec 2025
Abstract
Liver transplantation (LT) is a curative treatment for hepatocellular carcinoma (HCC). However, lifelong immunosuppressive therapy required to prevent graft rejection inevitably compromises antitumor immunity, thereby increasing the risk of HCC recurrence and metastasis, particularly common in the lungs. This review delves into the [...] Read more.
Liver transplantation (LT) is a curative treatment for hepatocellular carcinoma (HCC). However, lifelong immunosuppressive therapy required to prevent graft rejection inevitably compromises antitumor immunity, thereby increasing the risk of HCC recurrence and metastasis, particularly common in the lungs. This review delves into the complex dynamic equilibrium between immune cell subsets mediating rejection and antitumor immunity, systematically analyzes the impact of current immunosuppressive regimens on this balance, and highlights emerging strategies aimed at minimizing rejection while preserving or enhancing antitumor efficacy. These strategies include immunosuppressive regimen optimization, such as mTOR inhibitor application and calcineurin inhibitor (CNI) minimization, novel immunotherapies, including immune checkpoint inhibitors (ICIs) and adoptive cell therapy (ACT), and immune tolerance induction. This review also summarizes advances in biomarker research guiding immunosuppressant withdrawal, aiming to provide a theoretical basis and clinical insights for personalized immunotherapy strategies and comprehensive tumor management in LT recipients with HCC. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

19 pages, 6498 KB  
Article
Dihydromyricetin Remodels the Tumor Immune Microenvironment in Hepatocellular Carcinoma: Development and Validation of a Prognostic Model
by Yang Xu, Chao Gu, Wei Li, Fei Lan, Jingkun Mao, Xiao Tan and Pengfei Li
Curr. Issues Mol. Biol. 2025, 47(12), 1010; https://doi.org/10.3390/cimb47121010 - 2 Dec 2025
Abstract
Background: Dihydromyricetin (DHM), a natural dihydroflavonol, exhibits diverse pharmacological properties, including anti-inflammatory, antioxidant, and anti-tumor effects. However, its potential mechanism of action in the individualized therapy of hepatocellular carcinoma (HCC) remains unclear. Methods: Potential therapeutic targets of DHM were identified using the Swiss [...] Read more.
Background: Dihydromyricetin (DHM), a natural dihydroflavonol, exhibits diverse pharmacological properties, including anti-inflammatory, antioxidant, and anti-tumor effects. However, its potential mechanism of action in the individualized therapy of hepatocellular carcinoma (HCC) remains unclear. Methods: Potential therapeutic targets of DHM were identified using the Swiss Target Prediction database. The overlap between these targets and differentially expressed genes in HCC was analyzed to determine therapeutic targets. A prognostic model was constructed based on these genes, and patients were stratified into high- and low-risk groups. The associations between risk scores, clinical pathological characteristics, and overall survival were analyzed using Cox regression and Kaplan–Meier survival curves. The relationships between risk score and immune cell infiltration, immunosuppressive factors, and anticancer drug susceptibility were evaluated. Results: A three-gene prognostic model was established, comprising DTYMK, MAPT, and UCK2, designated as DHM-target genes (DHMGs). Patients in the high-risk group had significantly shorter overall survival than those in the low-risk group (p < 0.001; HR [95% CI] = 4.953 [2.544, 9.645]). Higher risk scores were correlated with more advanced tumor stages and grades. Comprehensive analysis of the tumor immune microenvironment revealed that high-risk patients exhibited significantly elevated TIDE scores, increased Treg cell infiltration, and markedly reduced stromal scores. Conclusions: This study developed a prognostic model based on the potential target genes of DHM in HCC. This model effectively stratifies HCC patients, identifying a high-risk subgroup characterized by an immunosuppressive microenvironment. These findings provide a theoretical foundation for exploring DHM as a promising natural adjuvant for cancer immunotherapy. Full article
(This article belongs to the Special Issue Challenges and Advances in Bioinformatics and Computational Biology)
Show Figures

Figure 1

24 pages, 3261 KB  
Case Report
Fatal Myocarditis Following Adjuvant Immunotherapy: A Case Report and Literature Review
by Nanteznta Torounidou, Melina Yerolatsite, Vasileios Bouratzis, Anna-Lea Amylidi, George Zarkavelis, Katerina K. Naka, Paraskevi V. Voulgari and Stergios Boussios
Int. J. Mol. Sci. 2025, 26(23), 11646; https://doi.org/10.3390/ijms262311646 - 1 Dec 2025
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy by enhancing the immune response against tumors. However, they can cause immune-related adverse events (irAEs), including rare but potentially fatal myocarditis. We describe a 71-year-old man with stage IIIA lung adenocarcinoma treated with adjuvant pembrolizumab [...] Read more.
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy by enhancing the immune response against tumors. However, they can cause immune-related adverse events (irAEs), including rare but potentially fatal myocarditis. We describe a 71-year-old man with stage IIIA lung adenocarcinoma treated with adjuvant pembrolizumab who developed severe ICI-associated myocarditis. Despite early diagnosis, treatment with intensive immunosuppression and mechanical support, he suffered fatal cardiac complications. A systematic review of the literature up to May 2025 identified 44 cases of ICI-associated myocarditis. Data on clinical features, diagnostics, treatment, and outcomes were extracted and analyzed. Most cases involved older patients with lung cancer treated with pembrolizumab or nivolumab. Onset varied from days to years after therapy initiation. Presentations included dyspnea, chest pain, arrhythmias, and elevated cardiac biomarkers. The biopsy showed T-cell and macrophage infiltration. High-dose corticosteroids were the primary treatment; additional immunosuppressants were used in cases that were refractory. Mortality was 45%, mainly due to cardiac failure and sepsis. Discussion: ICI-associated myocarditis arises from immune dysregulation affecting cardiac tissue, potentially involving shared antigens and systemic inflammation. Early detection and aggressive immunosuppression are crucial but often insufficient, resulting in high mortality. This underscores the urgent need for a better understanding of pathogenesis and the development of effective management strategies to improve patient outcomes. Finally, a multidisciplinary approach is important to improve outcomes in ICI-associated myocarditis. Full article
Show Figures

Figure 1

23 pages, 328 KB  
Review
An Update on Dermatomyositis and Related Inflammatory Myopathies: Cutaneous Clues, Skeletal Muscle Involvement, and Advances in Pathogenesis and Treatment
by Andres Parga, Dhruv Ratra and Dana Luu
Muscles 2025, 4(4), 58; https://doi.org/10.3390/muscles4040058 (registering DOI) - 1 Dec 2025
Abstract
Dermatomyositis (DM) is a prototypic idiopathic inflammatory myopathy in which characteristic skin disease frequently precedes or parallels muscle involvement and signals risks such as interstitial lung disease (ILD) and malignancy. This literature review integrates recent advances across dermatology, neuromuscular medicine, and immunology to [...] Read more.
Dermatomyositis (DM) is a prototypic idiopathic inflammatory myopathy in which characteristic skin disease frequently precedes or parallels muscle involvement and signals risks such as interstitial lung disease (ILD) and malignancy. This literature review integrates recent advances across dermatology, neuromuscular medicine, and immunology to refine diagnosis and management. We surveyed the literature from 2000 to 2025, prioritizing randomized trials, large cohorts, and translational studies that spanned classic and juvenile DM, amyopathic/hypomyopathic variants, and overlap phenotypes. Key insights include the diagnostic weight of pathognomonic cutaneous lesions with nailfold microangiopathy; the utility of myositis-specific autoantibodies for endotyping and risk (e.g., anti-TIF1-γ/anti-NXP2 and cancer, anti-MDA5 and rapidly progressive ILD); and the value of myxovirus-resistance protein A (MxA) immunohistochemistry and muscle MRI patterning (including distinctions from immune-mediated necrotizing myopathy) when enzymes are normal, or biopsies are treatment-modified. Management is anchored in early steroid-sparing immunosuppression tailored to phenotype, with evidence for IVIG in active DM and growing support for JAK inhibition, particularly in interferon-high or anti-MDA5 ILD, alongside selective use of calcineurin inhibitors and rituximab, with plasma exchange considered for refractory, rapidly progressive ILD. We highlight risk-stratified malignancy screening (IMACS 2023) and complications, including calcinosis, lipodystrophy, and chronic cutaneous damage. Skin-led recognition coupled with antibody-guided, phenotype-directed therapy and interdisciplinary care offers a pragmatic precision framework to improve outcomes and reduce long-term disability. Full article
19 pages, 295 KB  
Article
Factors Associated with Candidemia After Living Donor Liver Transplantation: A Case–Control Study
by Mefkure Durmus, Sena Guzel Karahan, Sami Akbulut, Zeynep Burcin Yilmaz and Ertugrul Karabulut
J. Clin. Med. 2025, 14(23), 8516; https://doi.org/10.3390/jcm14238516 (registering DOI) - 1 Dec 2025
Abstract
Background: Liver transplant recipients are highly susceptible to invasive fungal infections, particularly candidemia, due to intensive immunosuppressive therapy and postoperative complications. However, few studies have comprehensively examined postoperative antimicrobial and immunosuppressive factors in this context. Aim: This study aimed to identify [...] Read more.
Background: Liver transplant recipients are highly susceptible to invasive fungal infections, particularly candidemia, due to intensive immunosuppressive therapy and postoperative complications. However, few studies have comprehensively examined postoperative antimicrobial and immunosuppressive factors in this context. Aim: This study aimed to identify perioperative and postoperative factors associated with the development of candidemia in living donor liver transplant (LDLT) recipients, with a particular focus on antimicrobial and immunosuppressive regimens during initial hospitalization. Methods: A retrospective case–control analysis was conducted involving 36 LDLT recipients who developed candidemia (candidemia group) and 72 matched controls without candidemia (non-candidemia group) between January 2019 and November 2023. Demographic and clinical variables were compared using univariate and multivariate logistic regression analyses to identify independent associations. A post hoc power analysis demonstrated a high statistical power (97.3%) to detect large effect sizes. Results: Univariate analysis revealed significant associations with prolonged intubation (p < 0.001), bile leaks (p < 0.001), relaparotomy (p < 0.001), chronic renal disease (p = 0.011), hepatocellular carcinoma (p = 0.011), and the use of antimicrobials including meropenem (p = 0.048), linezolid (p = 0.005), tigecycline (p = 0.045), third-generation cephalosporins (p = 0.003), anidulafungin (p < 0.001), fluconazole (p = 0.006), mycophenolate (p = 0.011), and total parenteral nutrition (TPN) (p = 0.049). CMV prophylaxis (p < 0.001) and CMV-PCR positivity (p = 0.015) were also significantly associated with candidemia. Multivariate logistic regression analysis identified prolonged intubation (OR = 1.07; p = 0.019), bile leaks (OR = 10.9; p = 0.002), anidulafungin use (OR = 4.70; p = 0.032), fluconazole use (OR = 35.8; p = 0.005), and absence of CMV prophylaxis (OR = 11.7; p = 0.021) as independent factors associated with increased odds of candidemia. Conclusions: Prolonged intubation, bile leaks, antifungal exposure, and lack of CMV prophylaxis are independently associated with higher odds of candidemia after LDLT. Targeted prophylaxis, prudent antimicrobial stewardship, and timely biliary intervention may reduce fungal morbidity and mortality in post-transplant patients. Full article
(This article belongs to the Section General Surgery)
13 pages, 533 KB  
Review
Peptide Vaccines for Pediatric High-Grade Glioma and Diffuse Midline Glioma: Current Progress and Future Perspectives
by Aron K. Mebrahtu, Vatsal Jain, Eliese M. Moelker, Alexandra M. Hoyt-Miggelbrink, Katayoun Ayasoufi and Eric M. Thompson
Vaccines 2025, 13(12), 1215; https://doi.org/10.3390/vaccines13121215 - 30 Nov 2025
Abstract
High-grade gliomas (HGGs) and diffuse midline gliomas (DMGs) in pediatric patients carry a poor prognosis, necessitating the rapid development of novel therapies. Peptide vaccines represent a safe, repeatable, and rational immunotherapeutic modality aimed at inducing potent, tumor-specific T-cell responses. In this review, we [...] Read more.
High-grade gliomas (HGGs) and diffuse midline gliomas (DMGs) in pediatric patients carry a poor prognosis, necessitating the rapid development of novel therapies. Peptide vaccines represent a safe, repeatable, and rational immunotherapeutic modality aimed at inducing potent, tumor-specific T-cell responses. In this review, we define the scope of current progress by arguing that immunogenicity in children with HGG/DMG hinges on three factors: appropriate antigen class (neoantigen vs. TAA), the use of potent immunoadjuvants, and successful navigation of immune suppression. To address the gap between biological promise and clinical reality, we analyze clinical trials targeting shared tumor-associated antigens (e.g., CMV pp65, Survivin) and specific shared neoantigens (H3.3K27M). Crucially, we highlight pivotal data from the PNOC007 trial, where the magnitude of H3.3K27M-specific T-cell expansion correlated directly with significantly longer overall survival (OS), establishing a causal link between pharmacodynamics and clinical benefit. However, the unique challenges of the immunosuppressive tumor microenvironment and the detrimental effect of necessary corticosteroids remain paramount barriers. Future success relies on multi-modal combination strategies, the development of next-generation personalized neoantigen vaccines, and the application of advanced neuroimaging to accurately assess treatment response. Full article
(This article belongs to the Special Issue The Development of Peptide-Based Vaccines)
Show Figures

Figure 1

16 pages, 1008 KB  
Review
Not So Benign: Revisiting Pure Membranous Lupus Nephritis
by Martina Uzzo, Marta Calatroni and Gabriella Luisa Moroni
J. Pers. Med. 2025, 15(12), 580; https://doi.org/10.3390/jpm15120580 (registering DOI) - 30 Nov 2025
Abstract
Pure membranous lupus nephritis (pMLN, ISN/RPS-class V) is a rare form of lupus nephritis (LN). Despite being associated with significant comorbidities, it has traditionally been considered a less aggressive subtype. Emerging data challenges this perception, highlighting its potential for chronic kidney disease progression [...] Read more.
Pure membranous lupus nephritis (pMLN, ISN/RPS-class V) is a rare form of lupus nephritis (LN). Despite being associated with significant comorbidities, it has traditionally been considered a less aggressive subtype. Emerging data challenges this perception, highlighting its potential for chronic kidney disease progression and kidney failure. pMLN is pathologically defined by subepithelial immune-complex deposits and typically presents with nephrotic syndrome, preserved renal function, and fewer systemic/immunologic manifestations compared to proliferative LN (ISN/RPS-classes III/IV). Repeat biopsies reveal frequent histological class switching from pMLN to proliferative and mixed LN forms, underscoring the dynamic nature of the disease and the limitations of clinical markers in reflecting histological activity. While the ISN/RPS kidney biopsy classification provides important prognostic insight, it does not fully capture underlying molecular heterogeneity. Recent advances in precision medicine, including proteomic and biomarker studies (e.g., EXT1/2, NCAM1), offer promising tools for patient stratification and tailored treatments. International guidelines now recommend immunosuppressive therapy for pMLN, aligning treatment strategies more closely with those for proliferative and mixed LN. Overall, pMLN should be considered a distinct but clinically relevant LN subtype requiring personalized management based on clinical, histological and molecular features. Long-term monitoring is essential, as baseline presentation does not reliably predict treatment response or disease trajectory. Full article
Show Figures

Figure 1

24 pages, 750 KB  
Review
Beyond Tumor Immunity: The Disruption of Endocrine and Infectious Homeostasis by Immune Checkpoint Inhibitors
by Ema Schönberger, Luka Švitek, Barbara Grubišić, Tara Cvijić Perić, Romana Marušić, Nika Vlahović Vlašić, Tomislav Kizivat, Silvija Canecki Varžić, Lorna Stemberger Marić and Ines Bilić Ćurčić
Int. J. Mol. Sci. 2025, 26(23), 11619; https://doi.org/10.3390/ijms262311619 - 30 Nov 2025
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by reactivating T cell-mediated anti-tumor immunity. However, this enhanced immune activity can lead to immune-related adverse events (irAEs). This narrative review focuses on endocrine irAEs, including thyroid dysfunction, hypophysitis, adrenal insufficiency, and diabetes mellitus. It [...] Read more.
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by reactivating T cell-mediated anti-tumor immunity. However, this enhanced immune activity can lead to immune-related adverse events (irAEs). This narrative review focuses on endocrine irAEs, including thyroid dysfunction, hypophysitis, adrenal insufficiency, and diabetes mellitus. It also explores infectious complications and their underlying mechanisms. These mechanisms include immune dysregulation resulting directly from ICI-induced T-cell activation and indirectly from the immunosuppressive therapies used to treat irAEs. Furthermore, potential role of endocrine irAEs in predisposing patients to infectious complications is analyzed. The objective is to provide non-oncology specialists with the clinical insight necessary to recognize and manage these complex side effects. This narrative review synthesizes current literature on the diagnosis, management, and pathophysiology of endocrine irAEs and infections associated with different classes of ICIs (anti-CTLA-4, anti-PD-1, and anti-PD-L1). Endocrine irAEs are common, with incidence varying by ICI type; combination therapies pose the highest risk. Thyroid dysfunction is the most frequent, followed by hypophysitis, which often leads to permanent secondary adrenal insufficiency. ICI-induced diabetes mellitus is a rare but serious complication, frequently presenting as diabetic ketoacidosis. ICIs are believed to induce a distinct array of infections resulting from immunological dysregulation, unrelated to immunosuppressive medication. The phenomenon is increasingly called ICI therapy-induced dysregulated immunity. Moreover, evidence suggests that endocrine irAEs can compromise immune function and lead to a significantly higher risk of bacterial and fungal infections. Identifying infections that imitate irAEs is particularly important because the therapy is significantly distinct. Greater interdisciplinary awareness is crucial for the early recognition and appropriate management of both the endocrine and infectious complications, ultimately improving the safety and outcomes for patients receiving immunotherapy. Full article
(This article belongs to the Special Issue Challenges of Immune Checkpoint Inhibitor Therapy)
Show Figures

Figure 1

15 pages, 1275 KB  
Review
Radiation-Induced Immune Responses from the Tumor Microenvironment to Systemic Immunity
by Shaun Png, Sirimuvva Tadepalli and Edward E. Graves
Cancers 2025, 17(23), 3849; https://doi.org/10.3390/cancers17233849 (registering DOI) - 30 Nov 2025
Abstract
Radiotherapy remains a central component of cancer therapy that induces DNA damage and cancer cell death. Beyond its cytotoxic effects, radiotherapy acts as a potent immunomodulator by releasing immunogenic molecules, inflammatory mediators, and neoantigens that shape anti-tumor immunity. Radiation-induced immune responses can have [...] Read more.
Radiotherapy remains a central component of cancer therapy that induces DNA damage and cancer cell death. Beyond its cytotoxic effects, radiotherapy acts as a potent immunomodulator by releasing immunogenic molecules, inflammatory mediators, and neoantigens that shape anti-tumor immunity. Radiation-induced immune responses can have opposing effects, including immune activation or immunosuppression that dictate local tumor responses. Increasing evidence suggests these immunologic effects are not confined to the site of irradiation, as radiation exposure to surrounding normal tissue can trigger systemic immune signaling that affects local tumor progression as well as metastatic spread. This review examines radiotherapy-induced immune responses across three interconnected contexts: (i) tumor-intrinsic signaling during radiation; (ii) tumor microenvironmental changes where innate and adaptive immune responses alter local outcomes; and (iii) systemic and off-target effects contributing to broader immune remodeling. A comprehensive understanding of radiotherapy-induced immune responses will guide therapeutic strategies to enhance its immunological potential while minimizing unintended immune and off-target effects. Full article
(This article belongs to the Special Issue Radiation Exposure, Inflammation and Cancers)
Show Figures

Figure 1

25 pages, 1984 KB  
Review
The Unnoticed Threat: Clinical Characteristics, Risk Factors, and Outcome of Mucormycosis in Solid Organ Transplantation
by Jorge Boán, Mario Fernández-Ruiz, Eduardo Aparicio-Minguijón and José María Aguado
J. Fungi 2025, 11(12), 853; https://doi.org/10.3390/jof11120853 (registering DOI) - 29 Nov 2025
Viewed by 57
Abstract
Mucormycosis is an uncommon but life-threatening invasive fungal infection caused by molds of the order Mucorales, whose incidence has increased among solid organ transplant (SOT) recipients in recent years. Profound immunosuppression (particularly high-dose corticosteroids), T-cell-depleting therapies, diabetes mellitus, and previous episodes of graft [...] Read more.
Mucormycosis is an uncommon but life-threatening invasive fungal infection caused by molds of the order Mucorales, whose incidence has increased among solid organ transplant (SOT) recipients in recent years. Profound immunosuppression (particularly high-dose corticosteroids), T-cell-depleting therapies, diabetes mellitus, and previous episodes of graft rejection are the main predisposing conditions. This narrative review summarizes the current evidence on epidemiology, pathogenesis, risk factors, clinical presentation, diagnostic strategies, and treatment outcomes of mucormycosis in the SOT population. Pulmonary and rhino-orbital-cerebral infections are the predominant clinical forms, often characterized by rapid angioinvasive progression and mortality rates exceeding 45%. Early diagnosis remains challenging due to nonspecific clinical manifestations and the limited sensitivity of conventional diagnostic tools, although molecular techniques such as the detection of circulating Mucorales DNA in blood and metagenomic next-generation sequencing are promising. Liposomal amphotericin B remains the first-line therapy, ideally associated to surgical debridement and reduction in immunosuppression, while broad-spectrum triazoles (isavuconazole and posaconazole) represent alternative or salvage options. Despite recent advances in diagnostic methods and antifungal therapy, the prognosis of post-transplant mucormycosis remains poor, underscoring the need for multidisciplinary management and collaborative studies to inform the clinical management in this high-risk population. Full article
(This article belongs to the Special Issue Mycological Research in Spain)
Show Figures

Figure 1

27 pages, 649 KB  
Review
Pulmonary Manifestations of Inborn Errors of Immunity: Diagnostic and Therapeutic Insights
by Katarzyna Napiorkowska-Baran, Szczepan Cofta, Paweł Treichel, Marta Tykwinska, Kinga Lis, Aleksandra Matyja-Bednarczyk, Bartłomiej Szymczak, Maciej Szota, Jozef Slawatycki, Michal Kulakowski and Zbigniew Bartuzi
Life 2025, 15(12), 1838; https://doi.org/10.3390/life15121838 - 29 Nov 2025
Viewed by 81
Abstract
Background: Inborn errors of immunity (IEIs) are a heterogeneous group of genetically determined disorders that lead to immune dysfunction, recurrent infections, and organ-specific complications. The lungs are among the most commonly affected organs, with both infectious and noninfectious manifestations that significantly contribute to [...] Read more.
Background: Inborn errors of immunity (IEIs) are a heterogeneous group of genetically determined disorders that lead to immune dysfunction, recurrent infections, and organ-specific complications. The lungs are among the most commonly affected organs, with both infectious and noninfectious manifestations that significantly contribute to morbidity and mortality. This study aimed to provide a comprehensive overview of pulmonary manifestations in IEI, with emphasis on pathophysiological mechanisms, diagnostic approaches, and therapeutic strategies. Methods: A narrative review and synthesis of current literature and clinical guidelines were conducted, focusing on pulmonary involvement in IEI as classified by the International Union of Immunological Societies (IUIS). The analysis included data on infectious and noninfectious complications, imaging findings, immunological assessments, and management strategies, supported by clinical evidence and expert consensus. Results: Pulmonary manifestations in IEI encompass a wide spectrum of conditions. Infectious complications include recurrent bacterial pneumonias, bronchitis, and opportunistic infections, frequently resulting in irreversible lung damage such as bronchiectasis. Noninfectious complications, including granulomatous–lymphocytic interstitial lung disease (GLILD) and interstitial lung disease (ILD), are common in disorders such as common variable immunodeficiency (CVID) and X-linked agammaglobulinemia (XLA). Early diagnosis using high-resolution computed tomography (HRCT) and immunological testing, combined with the timely initiation of immunoglobulin replacement therapy and anti-biotic prophylaxis, significantly improves prognosis. Conclusions: Pulmonary complications are key clinical indicators of IEI and often precede systemic manifestations. Early, integrated, and interdisciplinary diagnostic and therapeutic management are crucial for preventing irreversible lung damage and improving patient outcomes. Regular monitoring and individualized therapy, including immunoglobulin replacement, targeted immunosuppression, and vaccination, are the cornerstones of effective long-term care in IEI. Full article
(This article belongs to the Special Issue Treatment and Prognosis of Thoracic Diseases)
Show Figures

Figure 1

22 pages, 1001 KB  
Review
TCR-Based Antigen-Specific Therapy for Type 1 Diabetes Mellitus: From Editing Autoreactive Clones to Tolerance Induction
by Marina Fisher, Julia Philippova, Vasily Kurilin and Sergey Sennikov
Int. J. Mol. Sci. 2025, 26(23), 11563; https://doi.org/10.3390/ijms262311563 - 28 Nov 2025
Viewed by 32
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by the destruction of insulin-producing pancreatic β-cells by autoreactive T cells. Current treatments, including insulin replacement therapy and various immunotherapies, often modulate but fail to permanently halt the underlying autoimmune process or restore [...] Read more.
Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by the destruction of insulin-producing pancreatic β-cells by autoreactive T cells. Current treatments, including insulin replacement therapy and various immunotherapies, often modulate but fail to permanently halt the underlying autoimmune process or restore β-cell function. In this review, we examine T cell receptor (TCR)-based treatment strategies for T1DM. We focus on two main approaches: selective elimination of pathogenic autoreactive T cell clones and induction of immune tolerance using TCR-modified regulatory T cells (TCR-Tregs). We describe key islet autoantigens, including post-translationally modified neoantigens such as fusion insulin peptides, which are crucial for identifying pathogenic TCRs. Next, we will review methodologies for TCR detection and TCR-Treg generation, highlighting their mechanisms of action and impact on various immune cells, including dendritic cells, B cells, and macrophages. We will also examine the potential of CD8+T cell regulatory cells (CD8+Tregs). Finally, we will discuss the future of TCR-based therapy, emphasizing the need to optimize TCR affinity, ensure Tregs’ stability, and develop combination therapies. TCR-based therapy represents a revolutionary approach to restoring immune tolerance in T1DM, providing high specificity and reducing the risk of systemic immuno-suppression compared to traditional treatments. Full article
Show Figures

Figure 1

15 pages, 509 KB  
Review
Sepsis and the Liver
by Eleni V. Geladari, Anastasia-Amalia C. Kalergi, Apostolos A. Evangelopoulos and Vasileios A. Sevastianos
Diseases 2025, 13(12), 388; https://doi.org/10.3390/diseases13120388 (registering DOI) - 28 Nov 2025
Viewed by 19
Abstract
Background/Objectives: Sepsis-associated liver injury (SALI) is a critical and often early complication of sepsis, defined by distinct hyper-inflammatory and immunosuppressive phases that shape patient phenotypes. Methods: Characterizing these phases establishes a foundation for immunomodulation strategies tailored to individual immune responses, as discussed subsequently. [...] Read more.
Background/Objectives: Sepsis-associated liver injury (SALI) is a critical and often early complication of sepsis, defined by distinct hyper-inflammatory and immunosuppressive phases that shape patient phenotypes. Methods: Characterizing these phases establishes a foundation for immunomodulation strategies tailored to individual immune responses, as discussed subsequently. Results: The initial inflammatory response activates pathways such as NF-κB and the NLRP3 inflammasome, leading to a cytokine storm that damages hepatocytes and is frequently associated with higher SOFA scores and a higher risk of 28-day mortality. Kupffer cells and infiltrating neutrophils exacerbate hepatic injury by releasing proinflammatory cytokines and reactive oxygen species, thereby causing cellular damage and prolonging ICU stays. During the subsequent immunosuppressive phase, impaired infection control and tissue repair can result in recurrent hospital-acquired infections and a poorer prognosis. Concurrently, hepatocytes undergo significant metabolic disturbances, notably impaired fatty acid oxidation due to downregulation of transcription factors such as PPARα and HNF4α. This metabolic alteration corresponds with worsening liver function tests, which may reflect the severity of liver failure in clinical practice. Mitochondrial dysfunction, driven by oxidative stress and defective autophagic quality control, impairs cellular energy production and induces hepatocyte death, which is closely linked to declining liver function and increased mortality. The gut-liver axis plays a central role in SALI pathogenesis, as sepsis-induced gut dysbiosis and increased intestinal permeability allow bacterial products, including lipopolysaccharides, to enter the portal circulation and further inflame the liver. This process is associated with sepsis-related liver failure and greater reliance on vasopressor support. Protective microbial metabolites, such as indole-3-propionic acid (IPA), decrease significantly during sepsis, removing key anti-inflammatory signals and potentially prolonging recovery. Clinically, SALI most commonly presents as septic cholestasis with elevated bilirubin and mild transaminase changes, although conventional liver function tests are insufficiently sensitive for early detection. Novel biomarkers, including protein panels and non-coding RNAs, as well as dynamic liver function tests such as LiMAx (currently in phase II diagnostics) and ICG-PDR, offer promise for improved diagnosis and prognostication. Specifying the developmental stage of these biomarkers, such as identifying LiMAx as phase II, informs investment priorities and translational readiness. Current management is primarily supportive, emphasizing infection control and organ support. Investigational therapies include immunomodulation tailored to immune phenotypes, metabolic and mitochondrial-targeted agents such as pemafibrate and dichloroacetate, and interventions to restore gut microbiota balance, including probiotics and fecal microbiota transplantation. However, translational challenges remain due to limitations of animal models and patient heterogeneity. Conclusion: Future research should focus on developing representative models, validating biomarkers, and conducting clinical trials to enable personalized therapies that modulate inflammation, restore metabolism, and repair the gut-liver axis, with the goal of improving outcomes in SALI. Full article
(This article belongs to the Section Gastroenterology)
Show Figures

Figure 1

26 pages, 4422 KB  
Review
From Pathways to Patients in Atopic Dermatitis: Advanced Systemic Therapies
by Alvaro Prados-Carmona, Husein Husein-ElAhmed, Francisco José Navarro-Triviño and Ricardo Ruiz-Villaverde
Int. J. Mol. Sci. 2025, 26(23), 11487; https://doi.org/10.3390/ijms262311487 - 27 Nov 2025
Viewed by 155
Abstract
Atopic dermatitis is the most prevalent chronic inflammatory skin disease, posing a significant individual and healthcare burden. Traditionally managed with topical agents and broad immunosuppressants, the treatment landscape has shifted significantly in recent years. This review explores the transition from immunopathogenic understanding to [...] Read more.
Atopic dermatitis is the most prevalent chronic inflammatory skin disease, posing a significant individual and healthcare burden. Traditionally managed with topical agents and broad immunosuppressants, the treatment landscape has shifted significantly in recent years. This review explores the transition from immunopathogenic understanding to personalized treatment strategies through advanced systemic therapies. We provide a thorough description of the current therapeutic arsenal, including approved monoclonal antibodies and Janus kinase (JAK) inhibitors, as well as experimental agents under clinical investigation. Key cytokines and receptors implicated in type 2 inflammation are explored alongside relevant intracellular signaling pathways. Special attention has been given to literature published from 2015 onwards. By synthesizing the latest scientific and clinical knowledge, this review aims to provide clinicians with practical guidance for navigating the evolving landscape of atopic dermatitis management and improving patient outcomes. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapeutic Targets in Skin Diseases)
Show Figures

Figure 1

Back to TopTop