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
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
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

Search Results (8,636)

Search Parameters:
Keywords = anti-E5 antibodies

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 767 KB  
Systematic Review
Autoantibodies and Molecular Mimicry in Alphavirus Chronic Arthritis: A Systematic Review
by Nosipho Zanele Masoto and Felicity Jane Burt
Pathogens 2026, 15(2), 152; https://doi.org/10.3390/pathogens15020152 - 30 Jan 2026
Abstract
Chronic arthritis following arthritogenic alphavirus infections presents symptoms resembling autoimmune rheumatic diseases, raising questions about the underlying mechanisms, including molecular mimicry and autoantibody production. This systematic review evaluated evidence supporting molecular mimicry and the potential role of autoantibodies as predictive biomarkers in alphavirus-induced [...] Read more.
Chronic arthritis following arthritogenic alphavirus infections presents symptoms resembling autoimmune rheumatic diseases, raising questions about the underlying mechanisms, including molecular mimicry and autoantibody production. This systematic review evaluated evidence supporting molecular mimicry and the potential role of autoantibodies as predictive biomarkers in alphavirus-induced chronic arthritis. A comprehensive search of PubMed, Scopus and Web of Science was conducted following PRISMA 2020 guidelines and PECO framework. Thirteen studies met the inclusion criteria: four computational studies assessing peptide homology between viral and human proteins, and nine clinical studies evaluating autoantibodies in chronic post-alphavirus arthritis. Computational analyses identified conserved alphavirus peptides with sequence and structural similarity to human proteins implicated in autoimmunity, supporting the hypothesis of molecular mimicry. However, most lacked experimental validation. Clinical studies showed variable detection of autoantibodies, rheumatoid factors, anti-cyclic citrullinated peptide, and antinuclear antibodies in chronic patients, though seropositivity rates were inconsistent and generally low. Only one study reported a significant association between autoantibody levels and disease chronicity. The findings suggest a potential autoimmune component in post-alphavirus arthritis driven by molecular mimicry, though current evidence remains inconclusive due to methodological heterogeneity and limited validation. Autoantibodies may contribute to pathogenesis but are not reliable predictors of chronicity. Future longitudinal studies with standardized assays and validation of computational findings in human models are needed. Full article
(This article belongs to the Special Issue Pathogen–Host Interactions: Death, Defense, and Disease)
12 pages, 1436 KB  
Article
Sensitive Visual Detection of Breast Cancer Cells via a Dual-Receptor (Aptamer/Antibody) Lateral Flow Biosensor
by Yurui Zhou, Jiahui Wang, Ying Han, Meijing Ma, Junhong Li, Haidong Li, Xueji Zhang and Guodong Liu
Biosensors 2026, 16(2), 85; https://doi.org/10.3390/bios16020085 - 30 Jan 2026
Abstract
We report a novel dual-receptor lateral flow biosensor (LFB) for the rapid, sensitive, and visual detection of MCF-7 breast cancer cells as a model for circulating tumor cells (CTCs). The biosensor employs a MUC1-specific aptamer conjugated to colloidal gold nanoparticles as the detection [...] Read more.
We report a novel dual-receptor lateral flow biosensor (LFB) for the rapid, sensitive, and visual detection of MCF-7 breast cancer cells as a model for circulating tumor cells (CTCs). The biosensor employs a MUC1-specific aptamer conjugated to colloidal gold nanoparticles as the detection probe and an anti-MUC1 antibody immobilized at the test line as the capture probe, forming a unique “aptamer–cell–antibody” sandwich complex upon target recognition. This design enables instrument-free, visual readout within minutes, achieving a detection limit of 675 cells. The assay also demonstrates robust performance in spiked human blood samples, highlighting its potential as a simple, cost-effective dual-mode point-of-care testing (POCT) platform. This platform supports both rapid visual screening and optional strip-reader-based quantification, making it suitable for early detection and monitoring of breast cancer CTCs. Full article
(This article belongs to the Special Issue The Research and Application of Lateral Flow Biosensors)
15 pages, 3755 KB  
Article
Inducible Costimulator and Its Ligand Promote Proliferation and Migration of Tumor Cells in Cutaneous T-Cell Lymphoma
by Kenta Oka, Takuya Miyagawa, Hiromichi Morita, Hiraku Suga, Tomomitsu Miyagaki, Sayaka Shibata, Hiroaki Kamijo, Yuka Mizuno, Teruyoshi Hisamoto, Issei Omori, Hikari Boki, Tomonori Oka, Naomi Takahashi-Shishido, Makoto Sugaya and Shinichi Sato
Int. J. Mol. Sci. 2026, 27(3), 1408; https://doi.org/10.3390/ijms27031408 - 30 Jan 2026
Abstract
Inducible costimulator (ICOS) is a costimulatory immune checkpoint receptor expressed on activated T-cells, while the ICOS ligand (ICOSL) is expressed on antigen-presenting cells. The ICOS–ICOSL axis promotes the survival of memory and effector T-cells and induces several immune responses. In addition, the ICOS–ICOSL [...] Read more.
Inducible costimulator (ICOS) is a costimulatory immune checkpoint receptor expressed on activated T-cells, while the ICOS ligand (ICOSL) is expressed on antigen-presenting cells. The ICOS–ICOSL axis promotes the survival of memory and effector T-cells and induces several immune responses. In addition, the ICOS–ICOSL interaction induces cell proliferation, cell survival, and cytokine production. The roles of ICOS and ICOSL in cutaneous T-cell lymphoma (CTCL) are unclear. In this study, we examined the roles of ICOS and ICOSL in CTCL. The tumor cells co-expressed ICOS and ICOSL, and the upregulated expression of ICOS and ICOSL reflected disease severity. Anti-ICOS and anti-ICOSL neutralizing antibodies inhibited both the in vitro and in vivo proliferation of CTCL cell lines. The anti-ICOSL neutralizing antibodies induced apoptosis and suppressed CCR4 expression on tumor cells, inhibiting CCR4–CCL17-mediated migration. These results suggest that the ICOS–ICOSL axis plays an essential role in CTCL pathogenesis, and targeting the ICOS–ICOSL axis could be a viable strategy for treating CTCL. Full article
Show Figures

Figure 1

10 pages, 763 KB  
Article
Correlation Analysis Between HLA Polymorphisms and Immune Response to Hepatitis B Vaccine in Children with Acute Lymphoblastic Leukemia
by Rui Zhang, Tian Yang, Yijin Gao, Hua Zhang, Yi Fei, Laibao Yang and Pengfei Deng
Vaccines 2026, 14(2), 145; https://doi.org/10.3390/vaccines14020145 - 30 Jan 2026
Abstract
Background: The human leukocyte antigen (HLA) is crucial for antigen presentation and vaccine efficacy. This study examined the association between HLA polymorphisms and the immune response to hepatitis B vaccination in children with acute lymphoblastic leukemia (ALL). Methods: 101 pediatric ALL patients at [...] Read more.
Background: The human leukocyte antigen (HLA) is crucial for antigen presentation and vaccine efficacy. This study examined the association between HLA polymorphisms and the immune response to hepatitis B vaccination in children with acute lymphoblastic leukemia (ALL). Methods: 101 pediatric ALL patients at Shanghai Children's Medical Center affiliated with Shanghai Jiaotong University School of Medicine who tested negative for hepatitis B surface antibody (anti-HBs) and were not infected with hepatitis B received three doses of the hepatitis B vaccine. Anti-HBs titers were measured before and after vaccination. Participants were divided into high- and low-response groups based on post-vaccination anti-HBs titers. Sequence-specific primer polymerase chain reaction (PCR-SSP) was used to genotype HLA-A, -B, -Cw, -DRB1, and -DQB1 alleles. Results: Pre-vaccination anti-HBs titers were 3.38 ± 2.97 mIU/mL, and the post-vaccination seroconversion rate was 100% with mean titers of 429.61 ± 303.13 mIU/mL (p < 0.001). Following immunization, the low-response group (11.88%) had an anti-HBs titer of 56.47 ± 28.38 mIU/mL, while the high-response group (88.12%) had an anti-HBs titer of 479.93 ± 287.70 mIU/mL. There were significant differences in allele frequencies of B*3501 and Cw*0303 between the two response groups (p < 0.05). Binary logistic regression analysis showed that the B*3501 allele was negatively correlated with the anti-HBs response level (p < 0.05). Conclusions: HLA-B*3501 may be associated with lower antibody response levels in children with ALL who completed the full hepatitis B vaccination series. All these children demonstrated protection against the hepatitis B virus (HBV). We will subsequently validate the association between HLA-B*3501 and the level of hepatitis B vaccine immune response in children with ALL through expanding the sample size or conducting a multicenter study. Full article
(This article belongs to the Section Hepatitis Virus Vaccines)
40 pages, 4302 KB  
Review
Immunotherapy Approaches for the Treatment of Triple-Negative Breast Cancer
by Shaimaa Alharbi, Farah Faozi Qasem, Mahsa Taleb Talebi, Nourhan E. Omran, Rifat Hamoudi and Rania Harati
Cancers 2026, 18(3), 464; https://doi.org/10.3390/cancers18030464 - 30 Jan 2026
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by high immunogenicity and specific immune signatures. Although these molecular features including elevated tumor-infiltrating lymphocytes (TILs) and programmed death-ligand 1 (PD-L1) expression provide a strong rationale for immunotherapy, clinical response remains [...] Read more.
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by high immunogenicity and specific immune signatures. Although these molecular features including elevated tumor-infiltrating lymphocytes (TILs) and programmed death-ligand 1 (PD-L1) expression provide a strong rationale for immunotherapy, clinical response remains limited due to multiple mechanisms of immune escape. This review summarizes current and emerging immunotherapeutic strategies in TNBC, including immune checkpoint inhibitors (PDL-1 and cytotoxic T-lymphocyte-associated protein 4 (CTL-4) blockade), adoptive cell therapy (ACT) (chimeric antigen receptor T-cell therapy (CAR-T) and TIL therapy), oncolytic virotherapy, and antibody-based approaches. We also discuss the mechanisms of resistance including DNA damage response alterations, anti-apoptotic signaling, and tumor microenvironment-mediated resistance. Finally, we highlight rational combination strategies, immunotherapy with chemotherapy, targeted therapy, or additional immunotherapies that aim to enhance response to immunotherapy. Ongoing advances in immunotherapy hold significant potential to improve outcomes for patients with TNBC. Full article
(This article belongs to the Special Issue Immunotherapy Approaches in Breast Cancer Treatment (2nd Edition))
13 pages, 1533 KB  
Article
A Real-World Experience of Cemiplimab in Patients with Advanced Cutaneous Squamous Cell Carcinoma
by Matteo Ravara, Tommaso Sani, Vincenzo D’Alonzo, Monica Valente, Elisa Cinotti, Clelia Miracco, Maura Colucci, Valentina Croce, Eleonora Carbonari, Ramiz Rana, Olindo Massarelli, Giovanni Rubino, Diana Giannarelli, Roberto Cuomo, Luca Grimaldi, Pietro Rubegni, Michele Maio and Anna Maria Di Giacomo
Cancers 2026, 18(3), 454; https://doi.org/10.3390/cancers18030454 - 30 Jan 2026
Abstract
Background: Cutaneous squamous cell carcinoma (cSCC) represents the second most common form of non-melanoma skin malignancy, and, when not amenable to curative surgery or radiotherapy, it is a life-threatening disease. The anti-PD-1 monoclonal antibody cemiplimab has transformed the outcome of advanced or metastatic [...] Read more.
Background: Cutaneous squamous cell carcinoma (cSCC) represents the second most common form of non-melanoma skin malignancy, and, when not amenable to curative surgery or radiotherapy, it is a life-threatening disease. The anti-PD-1 monoclonal antibody cemiplimab has transformed the outcome of advanced or metastatic cSCC, with response rates approaching 50% and sustained benefit beyond three years in clinical trials. Cemiplimab is now the first-line standard of care treatment for advanced disease. Methods: This retrospective observational study included consecutive adult patients with locally advanced (lac) or metastatic (m) cSCC who received cemiplimab (350 mg every three weeks) at the Center for Immuno-Oncology, University Hospital of Siena, Italy, either through an Expanded Access Program or routine clinical practice. Clinical outcome and treatment related adverse events (TRAEs) are reported. Results: Between December 2019 and December 2023, 27 patients (24 male; median age 82 years [range 41–90]) diagnosed with lacSCC (n = 20 [74.0%]) or mcSCC (n = 7 [25.9%]) were treated with cemiplimab as first line therapy and were followed until June 2024. Head and neck were the primary tumor location for 88.8% of patients, followed by trunk (7.4%) and lower extremities (3.7%). All patients had comorbidities, including six patients (22.2%) with hematologic malignancies. With a median follow-up of 31 months (data cut-off June 2024), the ORR was 66.6% (complete response 22.2%) and the disease control rate (DCR) 77.7%. Median progression-free survival (mPFS) and overall survival (mOS) were not reached, while 2-year PFS and OS rates were 65.2% and 71%, respectively. Treatment was well-tolerated, with three (11.1%) patients experiencing grade ≥3 TRAEs, and three patients discontinuing treatment due to TRAEs. Conclusions: Our real-world experience confirms the high rate of durable objective responses, good tolerability and long treatment duration of cemiplimab in elderly and frail cSCC patients as well. Full article
(This article belongs to the Special Issue Cancers in Dermatology—from Diagnosis to Treatment)
Show Figures

Figure 1

29 pages, 803 KB  
Review
Interleukins in COVID-19 and SARS-CoV-2 Variants: Immunopathogenesis, Therapeutic Perspectives and Vaccine-Induced Immune Responses
by Supriya Mahajan, Saurabh Mahajan and Akanksha Gusain
Int. J. Mol. Sci. 2026, 27(3), 1391; https://doi.org/10.3390/ijms27031391 - 30 Jan 2026
Abstract
The Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by profound immune dysregulation where interleukins play a central role in determining disease severity and response to interventions. This review summarizes the role of interleukins in the [...] Read more.
The Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by profound immune dysregulation where interleukins play a central role in determining disease severity and response to interventions. This review summarizes the role of interleukins in the immunopathogenesis of COVID-19, with particular emphasis on differences observed across major SARS-CoV-2 variants. Pro-inflammatory interleukins like IL-1β, IL-6, IL-2, IL-17 and IL-18 are critically involved in cytokine storm, hyperinflammation, and acute respiratory distress syndrome, whereas anti-inflammatory cytokines like IL-10 contribute to immune regulation and resolution of inflammation. Elevated levels of IL-1α, IL-1β, IL-4, IL-8, IL-9, IL-16, IL-18 have been documented in the Delta variant as compared with the Omicron variant, with IL-6 being the most frequent interleukin reported to be increased across all SARS-CoV-2 variants relative to the ancestral Wuhan strain. Elevated IL-2, IL-4, IL-6, and IL-10 levels have been associated with Omicron sub-variants. The review encompasses interleukin-based therapeutic strategies, where several IL-1 and IL-6 inhibitors were studied across clinical trials, but only tocilizumab has shown some promise against severe COVID-19. IL-2, IL-6, IL-15 and IL-21 levels were positively correlated with IgG and neutralizing antibody activity after vaccination with longevity of post-vaccination immunity being determined by IL-2 and IL-7. Full article
(This article belongs to the Special Issue Biochemistry and Molecular Biology of Coronaviruses)
Show Figures

Figure 1

18 pages, 439 KB  
Review
Neuroblastoma in Childhood: Biological Insights, Risk Stratification, and Advances in Multimodal Therapy
by Amina De Bona, Martina Barbieri, Nicole Rinaldi and Susanna Esposito
J. Clin. Med. 2026, 15(3), 1101; https://doi.org/10.3390/jcm15031101 - 30 Jan 2026
Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood and remains a leading cause of cancer-related mortality in pediatric patients. Characterized by marked clinical and biological heterogeneity, the disease ranges from spontaneously regressing tumors in infants to highly aggressive, treatment-resistant malignancies in [...] Read more.
Neuroblastoma is the most common extracranial solid tumor of childhood and remains a leading cause of cancer-related mortality in pediatric patients. Characterized by marked clinical and biological heterogeneity, the disease ranges from spontaneously regressing tumors in infants to highly aggressive, treatment-resistant malignancies in older children. Advances in molecular biology and genomics have significantly improved understanding of neuroblastoma pathogenesis, revealing the critical role of genetic and epigenetic alterations—such as MYCN amplification, ALK mutations, and chromosomal aberrations—in disease behavior and prognosis. Contemporary risk stratification systems now integrate clinical, biological, and molecular features to guide therapy more precisely. Management strategies have evolved toward risk-adapted, multimodal approaches. Low- and intermediate-risk patients often achieve excellent outcomes with surgery alone or limited chemotherapy, whereas high-risk neuroblastoma requires intensive multimodal treatment including induction chemotherapy, surgical resection, high-dose chemotherapy with autologous stem cell rescue, radiotherapy, and maintenance therapy. The incorporation of immunotherapeutic approaches, particularly anti-GD2 monoclonal antibodies, has significantly improved survival in high-risk disease. Emerging therapies such as targeted agents, radiopharmaceuticals, and cellular immunotherapies are further expanding the therapeutic landscape. Despite these advances, high-risk and relapsed neuroblastoma remain associated with substantial morbidity and mortality. Ongoing challenges include treatment resistance, long-term toxicity, and disparities in access to advanced therapies. Continued progress will depend on integrating molecular profiling into clinical decision-making, refining risk-adapted treatment strategies, and expanding international collaborative research efforts. This narrative review summarizes current knowledge on neuroblastoma epidemiology, biology, staging, and treatment, highlighting recent advances and future directions aimed at improving outcomes for affected children. Full article
Show Figures

Figure 1

19 pages, 2867 KB  
Article
Molecular Characterization of Persistent SARS-CoV-2 Infections in Immunocompromised Patients
by Patricia Volkow-Fernández, Marco Villanueva-Reza, Santiago Ávila-Ríos, Enrique Mendoza-Ramírez, América Citlali Vera-Jimenez, Alexandra Martin-Onraet, Beda Islas-Muñoz, Pamela Alatorre-Fernández, Rogelio Pérez-Padilla, Daniel Carpio-Guadarrama, Andrea Cárdenas-Ortega, Víctor Hugo Ahumada-Topete, Clara Espitia, Karen Lizbeth Reyes-Barrera, Edgar Sevilla-Reyes and Joel Armando Vázquez-Pérez
Viruses 2026, 18(2), 189; https://doi.org/10.3390/v18020189 - 30 Jan 2026
Abstract
Immunocompromised patients, including those with advanced HIV infection, hematologic malignancies treated with anti-CD20 monoclonal antibodies, or combined immunodeficiencies, are at increased risk of persistent SARS-CoV-2 infection. While long-term viral shedding has been described in these patients, the extent and nature of intra-host viral [...] Read more.
Immunocompromised patients, including those with advanced HIV infection, hematologic malignancies treated with anti-CD20 monoclonal antibodies, or combined immunodeficiencies, are at increased risk of persistent SARS-CoV-2 infection. While long-term viral shedding has been described in these patients, the extent and nature of intra-host viral evolution during long-term infection remain insufficiently documented. In this study, we report longitudinal genomic analyses of SARS-CoV-2 from three immunocompromised individuals with persistent COVID-19: (i) a female patient with follicular lymphoma receiving bendamustine-rituximab therapy with 9 months of persistence, (ii) a male patient with advanced HIV infection following prolonged antiretroviral therapy interruption with 10 months of persistence, and (iii) a female patient with Good’s Syndrome characterized by combined humoral and cellular immune deficiency with apparently four years of persistence. Replication-competent virus was detected over extended periods. Sequential whole-genome sequencing revealed the gradual accumulation of non-synonymous mutations across multiple viral genes, consistent with ongoing viral replication and intra-host diversification in the absence of effective immune control. Although based on a limited number of cases, these findings provide descriptive evidence that persistent SARS-CoV-2 infection in immunocompromised hosts can be associated with sustained viral evolution. This work highlights the importance of continued virological monitoring in selected patients with prolonged infection and contributes to the understanding of SARS-CoV-2 dynamics in settings of impaired immunity. Full article
(This article belongs to the Special Issue SARS-CoV-2, COVID-19 Pathologies, Long COVID, and Anti-COVID Vaccines)
Show Figures

Figure 1

13 pages, 1606 KB  
Article
Aptamers Targeting IL17A and Its Receptor Suppress IL17 Signaling in Different Cell Types
by Anastasiya Solovieva, Dariya Rippinen, Anna Davydova, Veronika Goncharova, Vladimir Koval, Mariya Vorobyeva and Maksim Korolev
Pharmaceuticals 2026, 19(2), 238; https://doi.org/10.3390/ph19020238 - 29 Jan 2026
Abstract
Background/Objectives: Interleukin-17A (IL-17A) is a key pathogenic cytokine in autoimmune arthropathies. Current monoclonal antibody inhibitors targeting the IL-17/IL-17RA axis demonstrate clinical efficacy but face significant limitations, including immunogenicity, the loss of therapeutic response, and cold-chain storage. Our study evaluated oligonucleotide aptamers targeting [...] Read more.
Background/Objectives: Interleukin-17A (IL-17A) is a key pathogenic cytokine in autoimmune arthropathies. Current monoclonal antibody inhibitors targeting the IL-17/IL-17RA axis demonstrate clinical efficacy but face significant limitations, including immunogenicity, the loss of therapeutic response, and cold-chain storage. Our study evaluated oligonucleotide aptamers targeting IL-17A and its receptor as an alternative to monoclonal antibodies to suppress an IL-17A-induced inflammatory response in cell models relevant to immunoinflammatory rheumatic diseases. Methods: We examined three aptamers: 2′-F-RNA aptamers Apt21-2 and Apt3-4 specific to IL-17A and DNA aptamer RA10-6 targeting the receptor of IL-17A. Their ability to suppress IL-17A functional activity was assessed in peripheral blood mononuclear cells (PBMCs) from healthy donors and personalized fibroblast-like synoviocytes (FLSs) from patients with axial spondyloarthritis (axSpA) and rheumatoid arthritis (RA). Inhibition was measured by quantifying IL-6 and MMP-13 secretion using ELISA and flow cytometry, using secukinumab as a reference control. Results: In PBMC, all aptamers suppressed IL-17A-stimulated IL-6 secretion and cell proliferation in a concentration-dependent manner (17–200 nM), with a 65–85% efficacy, comparable to that of secukinumab. In axSpA-derived FLS, we observed time-dependent efficacy: At 4 h, all three aptamers suppressed IL-6 to the same extent as secukinumab; at 24 h, RA10-6 maintained high efficacy while Apt21-2 and Apt3-4 showed reduced activity. A combination of receptor-targeting RA10-6 with anti-IL-17A aptamers resulted in synergistic IL-6 suppression. All aptamers reduced MMP-13 to basal levels. RA-derived FLS showed diminished responses to all inhibitors. Conclusions: Aptamers demonstrate high specificity and sustained efficacy in suppressing IL-17A signaling for an in vitro model of spondyloarthritis, with superior performance over antibodies. Disease-dependent differential efficacy in RA FLS reflects heterogeneity consistent with limited clinical anti-IL-17 efficacy in RA. These findings show the strong potential of the studied aptamers as an alternative to monoclonal antibodies for IL-17-associated inflammatory arthropathies, particularly spondyloarthritis. Full article
Show Figures

Figure 1

25 pages, 3229 KB  
Systematic Review
Major Advances in Gynecologic Oncology in 2025: Systematic Review and Synthesis of Conference and Published Evidence
by Nabil Ismaili
Biomedicines 2026, 14(2), 295; https://doi.org/10.3390/biomedicines14020295 - 28 Jan 2026
Abstract
Background: The year 2025 witnessed paradigm-shifting advances in gynecologic oncology, with pivotal clinical trial results redefining therapeutic standards across cervical, ovarian, endometrial, and vulvar cancers. Objectives: This systematic review aimed to comprehensively identify, synthesize, and critically evaluate pivotal phase II and [...] Read more.
Background: The year 2025 witnessed paradigm-shifting advances in gynecologic oncology, with pivotal clinical trial results redefining therapeutic standards across cervical, ovarian, endometrial, and vulvar cancers. Objectives: This systematic review aimed to comprehensively identify, synthesize, and critically evaluate pivotal phase II and III randomized controlled trials and major studies presented at the major annual meetings, alongside significant peer-reviewed publications from 2025 that introduce innovative therapeutic strategies across gynecologic malignancies. Methods: Conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 guidelines, this review involved exhaustive searches of electronic databases (PubMed/MEDLINE, Embase), conference proceedings (ASCO 2025, ESMO 2025), and major oncology journals for records from January to December 2025. Inclusion criteria encompassed: (1) Phase II or III randomized controlled trials (RCTs) and (2) Non-randomized studies (including phase I and II trials), reporting on novel therapeutic approaches in gynecologic oncology. All studies were required to report primary survival endpoints (overall survival or progression-free survival) or key efficacy outcomes. Study selection, data extraction, and methodological quality assessment were performed independently by two reviewers, with disagreements resolved through consensus or third-party adjudication. Results: From 1842 records, 23 studies met inclusion criteria (17 phase-III RCTs and 6 non-phase III RCTs/early-phase studies), distributed as follows: cervical cancer (9 studies, 39%), ovarian cancer (9 studies, 39%), endometrial cancer (4 studies, 17.5%), and vulvar cancer (1 study, 4.5%). The major advances identified include: (1) In cervical cancer, the KEYNOTE-A18 trial established pembrolizumab combined with chemoradiotherapy as a new standard for high-risk locally advanced disease, while the PHENIX trial validated sentinel lymph node biopsy as a safe surgical de-escalation strategy. (2) In ovarian cancer, the ENGOT-ov65/KEYNOTE-B96 trial demonstrated the first statistically significant overall survival improvement with an immune checkpoint inhibitor in platinum-resistant recurrent disease, establishing pembrolizumab plus weekly paclitaxel as a new standard of care. Novel therapeutic mechanisms, including glucocorticoid receptor modulation (ROSELLA trial) and cadherin-6-targeted antibody-drug conjugates (REJOICE-Ovarian01), showed remarkable efficacy. (3) In endometrial cancer, updated analyses from NRG GY018 and RUBY trials solidified the role of first-line immuno-chemotherapy, with differential benefits according to mismatch repair status. (4) In vulvar cancer, a pivotal phase II study demonstrated meaningful clinical activity of anti-PD-1 therapy in advanced disease. (5) The extensive circulating tumor DNA analysis from the CALLA trial provided crucial insights into biomarker dynamics in cervical cancer. Conclusions: The convergence of high-impact data from 2025 established multiple new standards of care, emphasizing biomarker-driven approaches, immunotherapy integration across disease stages, and novel mechanisms to overcome resistance, while highlighting challenges in treatment sequencing and global access. Full article
(This article belongs to the Special Issue Women’s Special Issue Series: Biomedicines (2nd Edition))
Show Figures

Graphical abstract

13 pages, 1561 KB  
Article
Immunogenicity Evaluation of Six Key Antigens of Mycoplasma pneumoniae in BALB/c Mice
by Huang Luxia, Zhang Heng, Xiao Hongjian, Li Zhihua and Li Qianqian
Vaccines 2026, 14(2), 133; https://doi.org/10.3390/vaccines14020133 - 28 Jan 2026
Viewed by 22
Abstract
Background/Objective: Mycoplasma pneumoniae (MP) remains a significant pathogen causing respiratory tract inflammation, particularly in the elderly and children under 5 years old. The lack of an effective vaccine is primarily attributed to the absence of well-defined immunological targets. This study systematically evaluated [...] Read more.
Background/Objective: Mycoplasma pneumoniae (MP) remains a significant pathogen causing respiratory tract inflammation, particularly in the elderly and children under 5 years old. The lack of an effective vaccine is primarily attributed to the absence of well-defined immunological targets. This study systematically evaluated the immunological characteristics of six key MP proteins to facilitate vaccine development. Methods: We constructed recombinant plasmids (pET30a-CARDS/P1/P40-90/P30/P116/MPN133), expressed them in Escherichia coli, and evaluated their immunogenicity and immune reactivity against MP infection in BALB/c mice. Results: Six proteins induced strong antibody responses. Sera from all protein-immunized groups exhibited MP growth inhibitory activity, with P116 and MPN133 showing more pronounced inhibitory effects. After MP challenge, lung histopathological findings demonstrated that, except for the P1 group, the lung pathological scores of the protein-immunized groups were lower than those of the PBS control group, and the lung injury in the CARDS, P116 and MPN133 groups was significantly alleviated. Conclusion: The six recombinant proteins demonstrate good immunogenicity. Among them, CARDS induces a strong primary antibody response, while P116 and MPN133 elicit potent anti-MP antibodies. In addition, CARDS, P116 and MPN133 significantly alleviate lung pathological damage (n = 6, p < 0.01), indicating that these three proteins have greater potential as MP vaccine targets among the six candidate proteins. Full article
35 pages, 5794 KB  
Article
Targeting Colorectal Cancer Stem Cells Through Inhibition of the Fibroblast Growth Factor Receptor 4 Pathway with a Novel Antibody
by Gessica Filocamo, Mariachiara Buccarelli, Armin Lahm, Mirko Brunetti, Chantal Paolini, Gabriele De Luca, Michele Signore, Giorgia Castellani, Alessandra Boe, Romina Alfonsi, Mauro Biffoni, Ruggero De Maria, Lucia Ricci-Vitiani, Christian Steinkühler and Paola Gallinari
Cancers 2026, 18(3), 418; https://doi.org/10.3390/cancers18030418 - 28 Jan 2026
Viewed by 40
Abstract
Background/Objectives: The progression and dissemination of CRC are heavily influenced by a subpopulation of tumor cells known as CSCs. This study aimed to identify novel protein membrane antigens expressed by colorectal CSCs and the consequent development of targeted therapies based on monoclonal antibodies [...] Read more.
Background/Objectives: The progression and dissemination of CRC are heavily influenced by a subpopulation of tumor cells known as CSCs. This study aimed to identify novel protein membrane antigens expressed by colorectal CSCs and the consequent development of targeted therapies based on monoclonal antibodies directed against the identified antigens. Methods: Integrated bioinformatics analyses were conducted using proprietary CSC gene expression profiles and public colon gene expression databases, leading to the identification of five plasma membrane proteins enriched in CSCs. Genetic immunization in rats was employed to generate monoclonal antibodies (mAbs) targeting these antigens. FGFR4 was prioritized due to its overexpression in colorectal tumors. Its function was characterized in vitro and in vivo through assays evaluating proliferation, colony formation, migration, and tumorigenicity. The anti-FGFR4 antibody 3B6 was selected based on its affinity and ability to inhibit FGFR4 signaling in CSCs. Its therapeutic potential was further assessed in xenograft models, and alterations in downstream signaling were analyzed via Western blot. Results: FGFR4 emerged as a key regulator of CRC CSC proliferation, migration, and tumorigenic capacity. The 3B6 antibody, a high-affinity FGFR4 binder, demonstrated robust in vitro inhibition of CSC features and significant antitumor effects in patient-derived xenograft models. Western blot analysis confirmed the modulation of FGFR4-driven signaling pathways, particularly those involved in epithelial–mesenchymal transition (EMT). Conclusions: This study successfully identified several CSC-selective membrane antigens that can become therapeutic targets in CRC. Among them, we focused on FGFR4 as a promising target and developed the anti-FGFR4 3B6 monoclonal antibody which offers potential for both diagnostic and therapeutic applications. Full article
(This article belongs to the Section Cancer Drug Development)
Show Figures

Figure 1

13 pages, 1106 KB  
Article
HER2/neu Expression in Low-Grade Serous Ovarian Carcinoma: A Pilot Two-Center Retrospective Study
by Alina Badlaeva, Aleksandra Asaturova, Aleksandra Rogozhina, Larisa Ezhova, Natalia Arzhanukhina, Anna Tregubova, Dmitry Rogozhin and Gennady Sukhikh
J. Clin. Med. 2026, 15(3), 1038; https://doi.org/10.3390/jcm15031038 - 28 Jan 2026
Viewed by 35
Abstract
Background/Objectives: Low-grade serous ovarian carcinoma (LGSOC) is known as an uncommon subtype of cancer with poor response to standard chemotherapy, so novel targets are required. The current study aims to highlight the Human Epidermal Growth Factor Receptor-2 (HER2/neu) immunohistochemical (IHC) expression in [...] Read more.
Background/Objectives: Low-grade serous ovarian carcinoma (LGSOC) is known as an uncommon subtype of cancer with poor response to standard chemotherapy, so novel targets are required. The current study aims to highlight the Human Epidermal Growth Factor Receptor-2 (HER2/neu) immunohistochemical (IHC) expression in LGSOCs. Methods: The study was conducted using 33 cases of LGSOCs from the calendar years 2017–2024. IHC staining was performed using antibody anti-HER2/neu (clone 4B5). HER2/neu scoring was performed based on the American Society of Clinical Oncology and the College of American Pathologists (ASCO/CAP) criteria for breast carcinoma. Results: The mean age of the 33 patients was 46.5 years. International Federation of Gynecology and Obstetrics (FIGO) stage data for patients revealed a predominance of advanced disease: 82.7% (24/29) were in advanced stages. Early stages comprised 17.3% (5/29) of cases. The study did not find HER2/neu overexpression in all cases. In 3.0% of samples (1/33), HER2/neu IHC staining was scored as 1+, and in 6.1% (2/33) of all LGSOCs, ultralow phenotype was observed. Of 23 cases in the HER2-negative group, 6 patients were alive with progressive disease, 1 patient died in 5 months, and 16 were alive with no evidence of disease. Of two patients with the HER2-ultralow phenotype, one was alive with no evidence of disease at 16 months follow-up. Conclusions: The results support the idea that HER2/neu overexpression is exceptionally rare in LGSOC; nevertheless, future trials are essential to fully characterize the spectrum of HER2/neu alterations in LGSOC and to determine definitively whether the rare cases with mutations or ultralow expression could represent a small subgroup that might benefit from specific targeted agents. Full article
Show Figures

Figure 1

17 pages, 1650 KB  
Article
Inductor-Based Biosensors for Real-Time Monitoring in the Liquid Phase
by Miriam Hernandez, Patricia Noguera, Nuria Pastor-Navarro, Marcos Cantero-García, Rafael Masot-Peris, Miguel Alcañiz-Fillol and David Gimenez-Romero
Biosensors 2026, 16(2), 79; https://doi.org/10.3390/bios16020079 - 28 Jan 2026
Viewed by 61
Abstract
Current liquid-phase resonant biosensors, such as Quartz Crystal Microbalance, Surface Acoustic Wave, or Surface Plasmon Resonance, typically rely on specialized piezoelectric substrates or complex optical setups. These requirements often necessitate cleanroom fabrication, thereby limiting cost-effective scalability. This study presents a high-integration sensing platform [...] Read more.
Current liquid-phase resonant biosensors, such as Quartz Crystal Microbalance, Surface Acoustic Wave, or Surface Plasmon Resonance, typically rely on specialized piezoelectric substrates or complex optical setups. These requirements often necessitate cleanroom fabrication, thereby limiting cost-effective scalability. This study presents a high-integration sensing platform based on standard Printed Circuit Board (PCB) technology, incorporating an embedded inductor within a fluidic system for real-time monitoring. This design leverages industrial manufacturing standards to achieve a compact, low-cost, and scalable architecture. Detection is governed by shifts in the resonance frequency of an LC tank circuit; specifically, increases in bulk ionic strength induce a frequency decrease, whereas biomolecular adsorption at the sensor surface leads to a frequency increase. This phenomenon can be explained by the modulation of the inter-turn capacitance, which is modeled as a combination of capacitive elements accounting for contributions from the bulk electrolyte and the surface-bound dielectric layer. Such divergent responses provide an intrinsic self-discriminating capability, allowing for the analytical differentiation between surface interactions and bulk effects. To the best of our knowledge, this is the first demonstration of an inductor-based resonant sensor fully embedded in a PCB fluidic architecture for continuous liquid-phase analyte monitoring. Validated through a protein-antibody model (Bovine Serum Albumin-anti-Bovine Serum Albumin), the sensor demonstrated a limit of detection of 1.7 ppm (0.026 mM) and a linear dynamic range of 31–211 ppm (0.47–3.2 mM). These performance metrics, combined with a reproducibility of 4 ± 3%, indicate that the platform meets the requirements for robust analytical applications. Its inherent simplicity and potential for miniaturization position this technology as a viable candidate for point-of-care diagnostics in diverse environments. Full article
(This article belongs to the Section Biosensor and Bioelectronic Devices)
Show Figures

Graphical abstract

Back to TopTop