Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (113)

Search Parameters:
Keywords = cytotoxic T lymphocytes resistance

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 1727 KiB  
Review
Immune Evasion in Head and Neck Squamous Cell Carcinoma: Roles of Cancer-Associated Fibroblasts, Immune Checkpoints, and TP53 Mutations in the Tumor Microenvironment
by Chung-Che Tsai, Yi-Chiung Hsu, Tin-Yi Chu, Po-Chih Hsu and Chan-Yen Kuo
Cancers 2025, 17(15), 2590; https://doi.org/10.3390/cancers17152590 - 7 Aug 2025
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive malignancy characterized by complex interactions within the tumor microenvironment (TME) that facilitate immune evasion and tumor progression. The TME consists of diverse cellular components, including cancer-associated fibroblasts, immune and endothelial cells, and [...] Read more.
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive malignancy characterized by complex interactions within the tumor microenvironment (TME) that facilitate immune evasion and tumor progression. The TME consists of diverse cellular components, including cancer-associated fibroblasts, immune and endothelial cells, and extracellular matrix elements, that collectively modulate tumor growth, metastasis, and resistance to therapy. Immune evasion in HNSCC is orchestrated through multiple mechanisms, including the suppression of cytotoxic T lymphocytes, recruitment of immunosuppressive cells, such as regulatory T and myeloid-derived suppressor cells, and upregulation of immune checkpoint molecules (e.g., PD-1/PD-L1 and CTLA-4). Natural killer (NK) cells, which play a crucial role in anti-tumor immunity, are often dysfunctional within the HNSCC TME due to inhibitory signaling and metabolic constraints. Additionally, endothelial cells contribute to tumor angiogenesis and immune suppression, further exacerbating disease progression. Recent advancements in immunotherapy, particularly immune checkpoint inhibitors and NK cell-based strategies, have shown promise in restoring anti-tumor immunity. Moreover, TP53 mutations, frequently observed in HNSCC, influence tumor behavior and therapeutic responses, highlighting the need for personalized treatment approaches. This review provides a comprehensive analysis of the molecular and cellular mechanisms governing immune evasion in HNSCC with a focus on novel therapeutic strategies aimed at improving patient outcomes. Full article
(This article belongs to the Special Issue Oral Cancer: Prevention and Early Detection (2nd Edition))
Show Figures

Figure 1

14 pages, 2113 KiB  
Article
NR2F6 as a Disease Driver and Candidate Therapeutic Target in Experimental Cerebral Malaria
by Victoria E. Stefan, Victoria Klepsch, Nikolaus Thuille, Martina Steinlechner, Sebastian Peer, Kerstin Siegmund, Peter Lackner, Erich Schmutzhard, Karin Albrecht-Schgör and Gottfried Baier
Cells 2025, 14(15), 1162; https://doi.org/10.3390/cells14151162 - 28 Jul 2025
Viewed by 265
Abstract
Cerebral malaria (CM) is the severe progression of an infection with Plasmodium falciparum, causing detrimental damage to brain tissue and is the most frequent cause of Plasmodium falciparum mortality. The critical role of brain-infiltrating CD8+ T cells in the pathophysiology of [...] Read more.
Cerebral malaria (CM) is the severe progression of an infection with Plasmodium falciparum, causing detrimental damage to brain tissue and is the most frequent cause of Plasmodium falciparum mortality. The critical role of brain-infiltrating CD8+ T cells in the pathophysiology of CM having been revealed, our investigation focuses on the role of NR2F6, an established immune checkpoint, as a candidate driver of CM pathology. We employed an experimental mouse model of CM based on Plasmodium berghei ANKA (PbA) infection to compare the relative susceptibility of Nr2f6-knock-out and wild-type C57BL6/N mice. As a remarkable result, Nr2f6 deficiency confers a significant survival benefit. In terms of mechanism, we detected less severe endotheliopathy and, hence, less damage to the blood–brain barrier (BBB), accompanied by decreased sequestered parasites and less cytotoxic T-lymphocytes within the brain, manifesting in a better disease outcome. We present evidence that NR2F6 deficiency renders mice more resistant to experimental cerebral malaria (ECM), confirming a causal and non-redundant role for NR2F6 in the progression of ECM disease. Consequently, pharmacological inhibitors of the NR2F6 pathway could be of use to bolster BBB integrity and protect against CM. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Figure 1

32 pages, 1691 KiB  
Review
Aptamers Targeting Immune Checkpoints for Tumor Immunotherapy
by Amir Mohammed Abker Abdu, Yanfei Liu, Rami Abduljabbar, Yunqi Man, Qiwen Chen and Zhenbao Liu
Pharmaceutics 2025, 17(8), 948; https://doi.org/10.3390/pharmaceutics17080948 - 22 Jul 2025
Viewed by 478
Abstract
Tumor immunotherapy has revolutionized cancer treatment by harnessing the immune system to recognize and eliminate malignant cells, with immune checkpoint inhibitors targeting programmed death receptor 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) demonstrating remarkable clinical success. However, challenges such [...] Read more.
Tumor immunotherapy has revolutionized cancer treatment by harnessing the immune system to recognize and eliminate malignant cells, with immune checkpoint inhibitors targeting programmed death receptor 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) demonstrating remarkable clinical success. However, challenges such as treatment resistance, immune-related adverse effects, and high costs highlight the need for novel therapeutic approaches. Aptamers, short, single-stranded oligonucleotides with high specificity and affinity for target molecules, have emerged as promising alternatives to conventional antibody-based therapies. This review provides a comprehensive analysis of aptamer-based strategies targeting immune checkpoints, with a particular focus on PD-1/PD-L1 and CTLA-4. We summarize recent advances in aptamer design, including bispecific and multifunctional aptamers, and explore their potential in overcoming immune resistance and improving therapeutic efficacy. Additionally, we discuss strategies to enhance aptamer stability, bioavailability, and tumor penetration through chemical modifications and nanoparticle conjugation. Preclinical and early clinical studies have demonstrated that aptamers can effectively block immune checkpoint pathways, restore T-cell activity, and synergize with other immunotherapeutic agents to achieve superior anti-tumor responses. By systematically reviewing the current research landscape and identifying key challenges, this review aims to provide valuable insights into the future directions of aptamer-based cancer immunotherapy, paving the way for more effective and personalized treatment strategies. Full article
(This article belongs to the Special Issue Nanomedicines for Overcoming Tumor Immunotherapy Tolerance)
Show Figures

Graphical abstract

22 pages, 17066 KiB  
Article
BST2 and DIRAS3 Drive Immune Evasion and Tumor Progression in High-Grade Glioma
by Zhangjun Liao, Shuyi Wu, Zhenyi Shi, Donghui Chen, Jinrui Chen and Hua Zhang
Int. J. Mol. Sci. 2025, 26(13), 6205; https://doi.org/10.3390/ijms26136205 - 27 Jun 2025
Viewed by 438
Abstract
High-grade gliomas (HGGs, WHO grades 3–4) are highly aggressive, with a poor prognosis and treatment resistance. Immune evasion may contribute to their progression, but the role of cytotoxic T-lymphocyte immune evasion (CTLE) is not well-validated. This study analyzed the transcriptomic data of 525 [...] Read more.
High-grade gliomas (HGGs, WHO grades 3–4) are highly aggressive, with a poor prognosis and treatment resistance. Immune evasion may contribute to their progression, but the role of cytotoxic T-lymphocyte immune evasion (CTLE) is not well-validated. This study analyzed the transcriptomic data of 525 patients from TCGA-GBM-HG_U133A. Two molecular subtypes were identified based on 182 CTLE-associated genes, with 238 differentially expressed genes between them. A prognostic model was developed, identifying BST2 and DIRAS3 as key risk factors, and validated in multiple cohorts. The subtypes had distinct immune profiles, with Cluster 2 showing higher immune infiltration but a poorer prognosis. The model had a good predictive performance. High-risk patients had upregulated BST2 and DIRAS3, linked to immunosuppression and shorter survival. Knockdown experiments confirmed their roles in GBM cell migration and invasion. Mechanistically, they promote immune evasion. BST2 and DIRAS3 could be therapeutic targets for HGG immunotherapy. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

21 pages, 1675 KiB  
Article
Ruxolitinib Modulates P-Glycoprotein Function, Delays T Cell Activation, and Impairs CCL19 Chemokine-Directed Migration in Human Cytotoxic T Lymphocytes
by Kipchumba Biwott, Algirmaa Lkhamkhuu, Nimrah Ghaffar, Albert Bálint Papp, Nastaran Tarban, Katalin Goda and Zsolt Bacso
Int. J. Mol. Sci. 2025, 26(13), 6123; https://doi.org/10.3390/ijms26136123 - 26 Jun 2025
Viewed by 750
Abstract
Ruxolitinib, a clinically approved JAK1/2 inhibitor used in the treatment of hematologic malignancies and inflammatory conditions, has been shown to interfere with the function of cytotoxic T lymphocytes (CTLs). Previous studies supported the involvement of the multidrug resistance transporter P-glycoprotein (Pgp/ABCB1) in CTL [...] Read more.
Ruxolitinib, a clinically approved JAK1/2 inhibitor used in the treatment of hematologic malignancies and inflammatory conditions, has been shown to interfere with the function of cytotoxic T lymphocytes (CTLs). Previous studies supported the involvement of the multidrug resistance transporter P-glycoprotein (Pgp/ABCB1) in CTL biology; however, the nature of its regulation remains unclear. To address this, we investigated the impact of ruxolitinib on Pgp expression and function in human CD8+ T cells. We demonstrate that CD8+ T lymphocytes express Pgp dynamically at both the mRNA and protein levels across naïve, short-term, and long-term activation states. Ruxolitinib increased the calcein accumulation in human Pgp-overexpressing NIH-3T3 cells and in CTLs and directly modulated Pgp function by increasing its basal ATPase activity in a concentration-dependent manner (10–100 μM), similar to the effect of the known Pgp substrate/modulator verapamil. Although measurable ATPase stimulation and transport inhibition were observed at supratherapeutic concentrations of ruxolitinib, its Pgp-mediated efflux may also occur at therapeutically relevant concentrations. In contrast, at therapeutically relevant plasma concentrations (1–3 μM), ruxolitinib significantly stabilized the mRNA expression of Pgp during early T-cell receptor (TCR) activation and inhibited the TCR-induced upregulation of Pgp, CD8, and PD-1 surface markers, suggesting its interference with activation-associated differentiation. At these same concentrations, ruxolitinib also impaired CCL19-directed transmigration of CTLs across human umbilical vein endothelial cell (HUVEC) monolayers, indicating disruption of lymphoid homing cues. Collectively, these findings demonstrate that ruxolitinib modulates Pgp at both the transcriptional and functional levels, with distinct concentration dependence. The ability of ruxolitinib to alter CTL activation and migration at clinically relevant plasma concentrations highlights the need for careful evaluation of JAK inhibitor–mediated immunomodulation and its implications for vaccination, transplantation, and T cell-based immunotherapies. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

18 pages, 2111 KiB  
Article
Consensus Sequences for Gag and Pol Introduced into HIV-1 Clade B Laboratory Strains Differentially Influence the Impact of Point Mutations Associated with Immune Escape and with Drug Resistance on Viral Replicative Capacity
by Sven Breitschwerdt, Benedikt Grandel, Benedikt Asbach, Franziska Winter, Todd Allen, Ralf Wagner, Bernd Salzberger and Arne Schneidewind
Viruses 2025, 17(6), 842; https://doi.org/10.3390/v17060842 - 12 Jun 2025
Viewed by 680
Abstract
Viral evasion from effective human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T-cell responses and from antiretroviral therapy through viral sequence variation is frequently accompanied by a loss in viral fitness. The impact of sequence variations on replication capacity in vitro was mostly studied [...] Read more.
Viral evasion from effective human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T-cell responses and from antiretroviral therapy through viral sequence variation is frequently accompanied by a loss in viral fitness. The impact of sequence variations on replication capacity in vitro was mostly studied by introducing single mutations into a specific clonal strain such as NL4-3. How the specific viral backbone itself impacts replicative fitness remains elusive. To test for a potential effect of the viral backbone, we constructed HIV-1 clade B clones with consensus sequences for gag and/or pol and evaluated the infectivity of viral variants harboring well-defined cytotoxic T-lymphocyte (CTL) escape mutations or drug resistance mutations within this backbone or the clonal NL4-3 strain. Viral variants with consensus sequences were replication-competent in vitro, although at lower rates than the NL4-3 virus. Introduction of the dominant CTL escape mutation R264K into the newly constructed viruses or into NL4-3 led to a dramatic reduction in infection rates. In contrast to the NL4-3 backbone, the combination of R264K with its compensatory mutation S173A on the consensus backbone led to higher infection rates as compared to the same virus in the absence of R264K and S173A. Furthermore, 2 out of 10 drug resistance mutations in pol led to opposing effects, with an increase in infection rates on the consensus gag/pol backbone and a reduction on NL4-3. Therefore, the effect of the respective viral backbone on infectivity observed in vitro might constitute an additional factor to explain differential kinetics of mutational evasion from immune and pharmaceutical pressure. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

26 pages, 705 KiB  
Review
Recent Advances in Molecular Research and Treatment for Melanoma in Asian Populations
by Soichiro Kado and Mayumi Komine
Int. J. Mol. Sci. 2025, 26(11), 5370; https://doi.org/10.3390/ijms26115370 - 3 Jun 2025
Viewed by 1083
Abstract
Melanoma treatment comprised a few treatment choices with insufficient efficacy before the emergence of molecularly targeted medication and immune checkpoint inhibitors, which dramatically improved patient outcomes. B-Rapidly Accelerated Fibrosarcoma (BRAF) and Mitogen-Activated Protein Kinase (MAPK) Kinase (MEK) inhibitors significantly improved survival in BRAF [...] Read more.
Melanoma treatment comprised a few treatment choices with insufficient efficacy before the emergence of molecularly targeted medication and immune checkpoint inhibitors, which dramatically improved patient outcomes. B-Rapidly Accelerated Fibrosarcoma (BRAF) and Mitogen-Activated Protein Kinase (MAPK) Kinase (MEK) inhibitors significantly improved survival in BRAF-mutant melanoma and immune checkpoint inhibitors, such as anti-programmed cell death 1 (PD-1) and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) agents, established new standards of care. Challenges remain, however, including the existence of resistance mechanisms and the reduced efficacy of immune-based therapies in Asian populations, particularly for acral and mucosal subtypes. This review highlights historical and current therapeutic advancements, discusses regional considerations, and explores emerging strategies aiming at globally optimizing melanoma management. Full article
Show Figures

Figure 1

24 pages, 1426 KiB  
Review
From Bench to Bladder: The Rise in Immune Checkpoint Inhibition in the Treatment of Non-Muscle Invasive Bladder Cancer
by Caitlin P. Burns, Jacob M. Parker, Dylan M. Schaap, Mark R. Wakefield and Yujiang Fang
Cancers 2025, 17(7), 1135; https://doi.org/10.3390/cancers17071135 - 28 Mar 2025
Cited by 1 | Viewed by 1035
Abstract
Non-muscle invasive bladder cancer (NMIBC) represents a significant clinical challenge due to its high recurrence rate and need for frequent monitoring. The current treatment modality is bacillus Calmette–Guérin (BCG) therapy combined with chemotherapy after transurethral resection of the bladder tumor (TURBT), which is [...] Read more.
Non-muscle invasive bladder cancer (NMIBC) represents a significant clinical challenge due to its high recurrence rate and need for frequent monitoring. The current treatment modality is bacillus Calmette–Guérin (BCG) therapy combined with chemotherapy after transurethral resection of the bladder tumor (TURBT), which is highly effective in most patients. Yet, the cancer becomes resistant to these treatments in 30–40% of patients, necessitating the need for new treatment modalities. In the cancer world, the development of immune checkpoint inhibitors that target molecules, such as programmed cell death protein-1 (PD-1), its ligand, PD-L1, and Cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), have revolutionized the treatment of many cancer types. PD-1/PD-L1 and CTLA-4 are shown to be upregulated in NMIBC in certain circumstances. PD-1/PD-L1 interactions play a role in immune evasion by suppressing T cell activity within the tumor microenvironment (TME), while the binding of CTLA-4 on T cells leads to downregulation of the immune response, making these pathways potential immunotherapeutic targets in NMIBC. This review seeks to understand the role of these therapies in treating NMIBC. We explore the cellular and non-cellular immune landscape in the TME of NMIBC, including Tregs, T effector cells, macrophages, B cells, and relevant cytokines. We also discuss the biological role of PD-1/PD-L1 and CTLA-4 while covering the rationale for these immunotherapies in NMIBC. Finally, we cover key clinical trials that have studied these treatments in NMIBC clinically. Such a study will be helpful for urologists and oncologists to manage patients with NMIBC more effectively. Full article
(This article belongs to the Special Issue Recent Advances in Non-muscle Invasive Bladder Cancer)
Show Figures

Figure 1

26 pages, 1653 KiB  
Review
The Role and Potential Application of IL-12 in the Immune Regulation of Tuberculosis
by Hangxing Wang, Guiren Ruan, Yuanchun Li and Xiaoqing Liu
Int. J. Mol. Sci. 2025, 26(7), 3106; https://doi.org/10.3390/ijms26073106 - 28 Mar 2025
Viewed by 1779
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant global health challenge, affecting millions annually and leading to substantial mortality, particularly in developing countries. The pathogen’s ability to persist latently and evade host immunity, combined with the emergence of drug-resistant [...] Read more.
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant global health challenge, affecting millions annually and leading to substantial mortality, particularly in developing countries. The pathogen’s ability to persist latently and evade host immunity, combined with the emergence of drug-resistant strains, underscores the need for innovative therapeutic strategies. This review highlights the crucial role of interleukin-12 (IL-12) in coordinating immune responses against TB, focusing on its potential as an immunotherapy target. IL-12, a key Th1 cytokine, enhances cellular immunity by promoting Th1 cell differentiation and IFN-γ production, vital for Mtb clearance. By stimulating cytotoxic T lymphocytes and establishing immune memory, IL-12 supports robust host defense mechanisms. However, the complexity of IL-12 biology, including its roles in pro-inflammatory and regulatory pathways, necessitates a nuanced understanding for effective therapeutic use. Recent studies have shown how IL-12 impacts T cell synapse formation, exosome-mediated bystander activation, and interactions with other cytokines in shaping T cell memory. Genetic defects in the IL-12/IFN-γ axis link to susceptibility to mycobacterial diseases, highlighting its importance in TB immunity. The review also addresses challenges like cytokine imbalances seen in TNF-α/IFN-γ synergy, which exacerbate inflammation, and the implications for IL-12-based interventions. Research into modulating IL-12, including its use as an adjuvant and in recombinant vaccines, promises improved TB treatment outcomes and vaccine efficacy. The review concludes by stressing the need for continued investigation into IL-12’s molecular mechanisms towards precision immunotherapies to combat TB and its complications. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Graphical abstract

51 pages, 2702 KiB  
Review
Advancing Breast Cancer Treatment: The Role of Immunotherapy and Cancer Vaccines in Overcoming Therapeutic Challenges
by Marco Palma
Vaccines 2025, 13(4), 344; https://doi.org/10.3390/vaccines13040344 - 24 Mar 2025
Cited by 1 | Viewed by 2430
Abstract
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body’s immune system to precisely target and eliminate cancer cells. However, several [...] Read more.
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body’s immune system to precisely target and eliminate cancer cells. However, several key factors influence the selection and effectiveness of these therapies, including BC subtype, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), PD-L1 expression, HER2 resistance, and the tumor microenvironment (TME). BC subtypes play a critical role in shaping treatment responses. Triple-negative breast cancer (TNBC) exhibits the highest sensitivity to immunotherapy, while HER2-positive and hormone receptor-positive (HR+) subtypes often require combination strategies for optimal outcomes. High TMB enhances immune responses by generating neoantigens, making tumors more susceptible to immune checkpoint inhibitors (ICIs); whereas, low TMB may indicate resistance. Similarly, elevated TIL levels are associated with better immunotherapy efficacy, while PD-L1 expression serves as a key predictor of checkpoint inhibitor success. Meanwhile, HER2 resistance and an immunosuppressive TME contribute to immune evasion, highlighting the need for multi-faceted treatment approaches. Current breast cancer immunotherapies encompass a range of targeted treatments. HER2-directed therapies, such as trastuzumab and pertuzumab, block HER2 dimerization and enhance antibody-dependent cellular cytotoxicity (ADCC), while small-molecule inhibitors, like lapatinib and tucatinib, suppress HER2 signaling to curb tumor growth. Antibody–drug conjugates (ADCs) improve tumor targeting by coupling monoclonal antibodies with cytotoxic agents, minimizing off-target effects. Meanwhile, ICIs, including pembrolizumab, restore T-cell function, and CAR-macrophage (CAR-M) therapy leverages macrophages to reshape the TME and overcome immunotherapy resistance. While immunotherapy, particularly in TNBC, has demonstrated promise by eliciting durable immune responses, its efficacy varies across subtypes. Challenges such as immune-related adverse events, resistance mechanisms, high costs, and delayed responses remain barriers to widespread success. Breast cancer vaccines—including protein-based, whole-cell, mRNA, dendritic cell, and epitope-based vaccines—aim to stimulate tumor-specific immunity. Though clinical success has been limited, ongoing research is refining vaccine formulations, integrating combination therapies, and identifying biomarkers for improved patient stratification. Future advancements in BC treatment will depend on optimizing immunotherapy through biomarker-driven approaches, addressing tumor heterogeneity, and developing innovative combination therapies to overcome resistance. By leveraging these strategies, researchers aim to enhance treatment efficacy and ultimately improve patient outcomes. Full article
(This article belongs to the Special Issue Advances in Cancer Immunotherapy and Vaccines Research: 2nd Edition)
Show Figures

Figure 1

24 pages, 1121 KiB  
Review
γδ T Cells: Game Changers in Immune Cell Therapy for Cancer
by Nabil Subhi-Issa, Daniel Tovar Manzano, Alejandro Pereiro Rodriguez, Silvia Sanchez Ramon, Pedro Perez Segura and Alberto Ocaña
Cancers 2025, 17(7), 1063; https://doi.org/10.3390/cancers17071063 - 21 Mar 2025
Cited by 1 | Viewed by 2382
Abstract
Gamma delta (γδ) T cells are a unique subset of T lymphocytes with distinctive features that make them highly promising candidates for cancer therapy. Their MHC-independent recognition of tumor antigens, ability to mediate direct cytotoxicity, and role in modulating the tumor microenvironment position [...] Read more.
Gamma delta (γδ) T cells are a unique subset of T lymphocytes with distinctive features that make them highly promising candidates for cancer therapy. Their MHC-independent recognition of tumor antigens, ability to mediate direct cytotoxicity, and role in modulating the tumor microenvironment position them as versatile agents in cancer immunotherapy. This review integrates and synthesizes the existing data on γδ T cells, with an emphasis on the development and optimization of in vitro expansion protocols. Critical aspects are detailed such as activation strategies, co-culture systems, cytokine use, and other parameters to ensure robust cell proliferation and functionality, which may be valuable for those developing or optimizing clinical practices. Finally, we discuss current advancements in γδ T cell research, clinical experience, and highlight areas needing further exploration. Considering these data, we hypothesize and propose potential new applications such as engineering γδ T cells for enhanced resistance to immune checkpoint pathways or for localized cytokine delivery within the tumor microenvironment, which could broaden their therapeutic impact in the treatment of cancer and beyond. Full article
(This article belongs to the Special Issue Advances in Molecular Oncology and Therapeutics)
Show Figures

Figure 1

17 pages, 1105 KiB  
Review
Programmed Cell Death Ligand as a Biomarker for Response to Immunotherapy: Contribution of Mass Spectrometry-Based Analysis
by Marco Agostini, Pietro Traldi and Mahmoud Hamdan
Cancers 2025, 17(6), 1001; https://doi.org/10.3390/cancers17061001 - 17 Mar 2025
Viewed by 772
Abstract
Immune checkpoint inhibition is a major component in today’s cancer immunotherapy. In recent years, the FDA has approved a number of immune checkpoint inhibitors (ICIs) for the treatment of melanoma, non-small-cell lung, breast and gastrointestinal cancers. These inhibitors, which target cytotoxic T-lymphocyte antigen-4, [...] Read more.
Immune checkpoint inhibition is a major component in today’s cancer immunotherapy. In recent years, the FDA has approved a number of immune checkpoint inhibitors (ICIs) for the treatment of melanoma, non-small-cell lung, breast and gastrointestinal cancers. These inhibitors, which target cytotoxic T-lymphocyte antigen-4, programmed cell death (PD-1), and programmed cell death ligand (PD-L1) checkpoints have assumed a leading role in immunotherapy. The same inhibitors exert significant antitumor effects by overcoming tumor cell immune evasion and reversing T-cell exhaustion. The initial impact of this therapy in cancer treatment was justly described as revolutionary, however, clinical as well as research data which followed demonstrated that these innovative drugs are costly, are associated with potentially severe adverse effects, and only benefit a small subset of patients. These limitations encouraged enhanced research and clinical efforts to identify predictive biomarkers to stratify patients who are most likely to benefit from this form of therapy. The discovery and characterization of this class of biomarkers is pivotal in guiding individualized treatment against various forms of cancer. Currently, there are three FDA-approved predictive biomarkers, however, none of which on its own can deliver a reliable and precise response to immune therapy. Present literature identifies the absence of precise predictive biomarkers and poor understanding of the mechanisms behind tumor resistance as the main obstacles facing ICIs immunotherapy. In the present text, we discuss the dual role of PD-L1 as a biomarker for response to immunotherapy and as an immune checkpoint. The contribution of mass spectrometry-based analysis, particularly the impact of protein post-translational modifications on the performance of this protein is underlined. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

11 pages, 658 KiB  
Review
Thyroid Cancer—The Tumor Immune Microenvironment (TIME) over Time and Space
by Juan Carlos Jaume
Cancers 2025, 17(5), 794; https://doi.org/10.3390/cancers17050794 - 26 Feb 2025
Viewed by 1295
Abstract
In thyroid cancer, the tumor immune microenvironment (TIME) plays a crucial role in cancer development, progression and response to treatment. Like many other cancers, thyroid cancer creates a complex network of interactions with immune cells directly (cell-to-cell) and via humoral mediators (i.e., cytokines). [...] Read more.
In thyroid cancer, the tumor immune microenvironment (TIME) plays a crucial role in cancer development, progression and response to treatment. Like many other cancers, thyroid cancer creates a complex network of interactions with immune cells directly (cell-to-cell) and via humoral mediators (i.e., cytokines). This dynamic microenvironment undergoes constant modification, which can lead to changes in the immunophenotype that might explain cancer progression, dedifferentiation and resistance to treatment. According to the cancer immunoediting hypothesis, cancerous tumors can shape their immune microenvironment to create an immunosuppressive milieu that allows them to evade classic immune surveillance. One mechanism by which this occurs is through the reprogramming of immune cells, often shifting their phenotypes from cytotoxic to regulatory. Recent research has shed light on cellular components and molecular interactions within the thyroid cancer TIME. Immune cells such as Tumor-Associated Lymphocytes (TALs), myeloid-derived suppressor cells (MDSCs), Tumor-Associated Macrophages (TAMs) and Double-Negative (DN) T cells seem to play key roles in shaping the immune response to thyroid cancer. Additionally, cytokines, chemokines and other signaling molecules contribute to the communication and regulation of immune cells within that microenvironment. By studying these interactions, researchers aim to uncover not just potential therapeutic targets but also biomarkers of thyroid cancer that could provide clues on severity and progression. Based on that knowledge, strategies such as the use of immune checkpoint inhibitors, antigen-specific targeted immunotherapies, and immunomodulatory agents are being explored to enhance the anti-tumor immune response and overcome cancer immunosuppressive mechanisms. In this review, we analyze the available literature and provide our own experience to unravel the complexity of the thyroid immune microenvironment. Continued research in this area holds promise for improving outcomes through the identification of immune markers of severity/progression of thyroid cancer and the development of innovative immunotherapeutic approaches. Full article
(This article belongs to the Special Issue Feature Papers in Section "Tumor Microenvironment")
Show Figures

Figure 1

16 pages, 2550 KiB  
Article
Computational and Population-Based HLA Permissiveness to HIV Drug Resistance-Associated Mutations
by Rizwan Mahmud, Zoë Krullaars, Jolieke van Osch, David Rickett, Zabrina L. Brumme, Kathryn S. Hensley, Casper Rokx, Rob A. Gruters, Jeroen J. A. van Kampen and Thibault Mesplède
Pathogens 2025, 14(3), 207; https://doi.org/10.3390/pathogens14030207 - 20 Feb 2025
Viewed by 939
Abstract
The presentation of HIV peptides by the human leukocyte antigen (HLA) complex to CD8+ cytotoxic T-cells (CTLs) is critical to limit viral pathogenesis. HIV can mutate to evade HLA-restricted CTL responses and resist antiretroviral drugs, raising questions about how it balances these evolutionary [...] Read more.
The presentation of HIV peptides by the human leukocyte antigen (HLA) complex to CD8+ cytotoxic T-cells (CTLs) is critical to limit viral pathogenesis. HIV can mutate to evade HLA-restricted CTL responses and resist antiretroviral drugs, raising questions about how it balances these evolutionary pressures. Here, we used a computational approach to assess how drug resistance-associated mutations (RAMs) affect the binding of HIV-1 subtype B or C peptides to the most prevalent HLA alleles in US, European, and South African populations. We predict RAMs that may be favored in certain populations and report the under-representation of Y181C in people expressing HLA-B*57:01. This finding agreed with our computational predictions when Y181C was at the major anchor site P2, suggesting the potential relevance of our approach. Overall, our findings lay out a conceptual framework to study the implications of HLA alleles on the emergence of HIV RAMs at the individual and population levels. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Figure 1

23 pages, 347 KiB  
Review
Advancements in Melanoma Treatment: A Review of PD-1 Inhibitors, T-VEC, mRNA Vaccines, and Tumor-Infiltrating Lymphocyte Therapy in an Evolving Landscape of Immunotherapy
by Apoorva Mehta, Mateen Motavaf, Ikenna Nebo, Sophia Luyten, Kofi D. Osei-Opare and Alejandro A. Gru
J. Clin. Med. 2025, 14(4), 1200; https://doi.org/10.3390/jcm14041200 - 12 Feb 2025
Cited by 4 | Viewed by 4081
Abstract
Melanoma, an aggressive skin cancer, presents significant therapeutic challenges. Consequently, innovative treatment strategies beyond conventional chemotherapy, radiation, and surgery are actively explored. This review discusses the evolution of immunotherapy in advanced melanoma, highlighting PD-1/PD-L1 inhibitors, mRNA vaccines, Talimogene Laherparepvec (T-VEC), and tumor-infiltrating lymphocyte [...] Read more.
Melanoma, an aggressive skin cancer, presents significant therapeutic challenges. Consequently, innovative treatment strategies beyond conventional chemotherapy, radiation, and surgery are actively explored. This review discusses the evolution of immunotherapy in advanced melanoma, highlighting PD-1/PD-L1 inhibitors, mRNA vaccines, Talimogene Laherparepvec (T-VEC), and tumor-infiltrating lymphocyte (TIL) therapies. PD-1/PD-L1 inhibitors such as pembrolizumab and nivolumab block immune checkpoints, promoting T-cell cytotoxic activity and improving overall survival in patients with advanced melanoma. T-VEC, a modified oncolytic herpes virus, promotes a systemic anti-tumor response while simultaneously lysing malignant cells. mRNA vaccines, such as Moderna’s mRNA-4157/V940, take advantage of malignant-cell-specific neoantigens to amplify the adaptive immune response while protecting healthy tissue. TIL therapy is a form of therapy involving ex vivo expansion and reinfusion of the patient’s tumor-specific lymphocytes and has been shown to provide durable tumor control. While these therapies have demonstrated promising clinical outcomes, challenges such as tumor resistance, high financial burden, and limited accessibility pose challenges to their widespread use. This review explores combination therapies such as PD-L1 inhibitors with mRNA vaccines, or TIL therapy, which aim to enhance treatment through synergistic approaches. Further research is required to optimize these combinations, address barriers preventing their use, and control adverse events. Full article
(This article belongs to the Section Dermatology)
Back to TopTop