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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (212)

Search Parameters:
Keywords = immunotherapy-related toxicity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 328 KiB  
Review
Adjuvant Immunotherapy in Stage IIB/IIC Melanoma: Current Evidence and Future Directions
by Ivana Prkačin, Ana Brkić, Nives Pondeljak, Mislav Mokos, Klara Gaćina and Mirna Šitum
Biomedicines 2025, 13(8), 1894; https://doi.org/10.3390/biomedicines13081894 - 4 Aug 2025
Viewed by 51
Abstract
Background: Patients with resected stage IIB and IIC melanoma are at high risk of recurrence and distant metastasis, despite surgical treatment. The recent emergence of immune checkpoint inhibitors (ICIs) has led to their evaluation in the adjuvant setting for early-stage disease. This [...] Read more.
Background: Patients with resected stage IIB and IIC melanoma are at high risk of recurrence and distant metastasis, despite surgical treatment. The recent emergence of immune checkpoint inhibitors (ICIs) has led to their evaluation in the adjuvant setting for early-stage disease. This review aims to synthesize current evidence regarding adjuvant immunotherapy for stage IIB/IIC melanoma, explore emerging strategies, and highlight key challenges and future directions. Methods: We conducted a comprehensive literature review of randomized clinical trials, observational studies, and relevant mechanistic and biomarker research on adjuvant therapy in stage IIB/IIC melanoma. Particular focus was placed on pivotal trials evaluating PD-1 inhibitors (KEYNOTE-716 and CheckMate 76K), novel vaccine and targeted therapy trials, mechanisms of resistance, immune-related toxicity, and biomarker development. Results: KEYNOTE-716 and CheckMate 76K demonstrated significant improvements in recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) with pembrolizumab and nivolumab, respectively, compared to placebo. However, no definitive overall survival benefit has yet been shown. Adjuvant immunotherapy is linked to immune-related adverse events, including permanent endocrinopathies. Emerging personalized approaches, such as circulating tumor DNA monitoring and gene expression profiling, may enhance patient selection, but remain investigational. Conclusions: Adjuvant PD-1 blockade offers clear RFS benefits in high-risk stage II melanoma, but optimal patient selection remains challenging, due to uncertain overall survival benefit and toxicity concerns. Future trials should integrate biomarker-driven approaches to refine therapeutic decisions and minimize overtreatment. Full article
(This article belongs to the Section Gene and Cell Therapy)
17 pages, 1474 KiB  
Review
Treatment Strategies for First-Line PD-L1-Unselected Advanced NSCLC: A Comparative Review of Immunotherapy-Based Regimens by PD-L1 Expression and Clinical Indication
by Blerina Resuli, Diego Kauffmann-Guerrero, Maria Nieves Arredondo Lasso, Jürgen Behr and Amanda Tufman
Diagnostics 2025, 15(15), 1937; https://doi.org/10.3390/diagnostics15151937 (registering DOI) - 31 Jul 2025
Viewed by 406
Abstract
Background: Lung cancer remains the leading cause of cancer-related mortality worldwide. Advances in screening, diagnosis, and management have transformed clinical practice, particularly with the integration of immunotherapy and target therapies. Methods: A systematic literature search was carried out for the period between October [...] Read more.
Background: Lung cancer remains the leading cause of cancer-related mortality worldwide. Advances in screening, diagnosis, and management have transformed clinical practice, particularly with the integration of immunotherapy and target therapies. Methods: A systematic literature search was carried out for the period between October 2016 to September 2024. Phase II and III randomized trials evaluating ICI monotherapy, ICI–chemotherapy combinations, and dual ICI regimens in patients with advanced NSCLC were included. Outcomes of interest included overall survival (OS), progression-free survival (PFS), and treatment-related adverse events (AEs). Results: PD-1-targeted therapies demonstrated superior OS compared to PD-L1-based regimens, with cemiplimab monotherapyranking highest for OS benefit (posterior probability: 90%), followed by sintilimab plus platinum-based chemotherapy (PBC) and pemetrexed—PBC. PFS atezolizumab plus bevacizumab and PBC, and camrelizumab plus PBC were the most effective regimens. ICI–chemotherapy combinations achieved higher ORRs but were associated with greater toxicity. The most favorable safety profiles were observed with cemiplimab, nivolumab, and avelumab monotherapy, while atezolizumab plus PBC and sugemalimab plus PBC carried the highest toxicity burdens. Conclusions: In PD-L1-unselected advanced NSCLC, PD-1 blockade—particularly cemiplimab monotherapy—and rationally designed ICI–chemotherapy combinations represent the most efficacious treatment strategies. Balancing efficacy with safety remains critical, especially in the absence of predictive biomarkers. These findings support a patient-tailored approach to immunotherapy and highlight the need for further biomarker-driven and real-world investigations to optimize treatment selection. Full article
(This article belongs to the Special Issue Lung Cancer: Screening, Diagnosis and Management: 2nd Edition)
Show Figures

Figure 1

22 pages, 1241 KiB  
Systematic Review
Safety and Efficacy of Immune Checkpoint Inhibitors in Human Immunodeficiency Virus-Associated Cancer: A Systematic Scoping Review
by Ahmed D. Alatawi, Amirah B. Alaqyl, Reema J. Alalawi, Rahaf S. Alqarni, Razan A. Sufyani, Ghadi S. Alqarni, Raghad S. Alqarni, Jumana H. Albalawi, Raghad A. Alsharif, Ghada I. Alatawi, Elaf N. Albalawi, Danah A. Alanazi, Sultanah A. Naitah, Reem Sayad and Helal F. Hetta
Diseases 2025, 13(8), 230; https://doi.org/10.3390/diseases13080230 - 22 Jul 2025
Viewed by 366
Abstract
Background/Objective: People living with human immunodeficiency virus (PHIV) are at increased risk for malignancies, yet their access to immunotherapy remains limited due to concerns about safety and efficacy. This systematic scoping review evaluates the use of immune checkpoint inhibitors (ICIs) in HIV-associated cancers, [...] Read more.
Background/Objective: People living with human immunodeficiency virus (PHIV) are at increased risk for malignancies, yet their access to immunotherapy remains limited due to concerns about safety and efficacy. This systematic scoping review evaluates the use of immune checkpoint inhibitors (ICIs) in HIV-associated cancers, analyzing patient outcomes, safety profiles, and the impact on HIV status. Methods: A comprehensive literature search was conducted in databases including PubMed, Scopus, Web of Science (WoS), and Medline, up to January 2025. Studies included assessing the efficacy of ICIs in cancer patients with HIV. The primary outcomes were (a) the efficacy of immune ICIs on prognosis, progression-free survival (PFS), and overall survival (OS). Secondary outcomes were the immune-related adverse events (irAEs) and the survival rate of cancer patients receiving ICIs. Results: A total of 107 cases from 19 studies published between 2011 and 2024 were reviewed. Responses to programmed death 1 (PD-1) inhibitors varied, with 27.1% achieving partial response, 23.36% experiencing stable disease, and 6.54% achieving complete response, while 34.57% had disease progression. Adverse events, including hematologic and endocrine toxicities, were common but mostly manageable. HIV viral loads remained stable in most cases. Conclusions: PD-1 inhibitors demonstrated potential efficacy in HIV-associated malignancies with a safety profile comparable to the general population. However, disease progression remained a concern, highlighting the need for optimized patient selection. Further well-controlled trials are essential to establish treatment guidelines and ensure equitable access to immunotherapy for PHIV. Full article
(This article belongs to the Special Issue Cancer Inhibitory Receptors and Related Cancer Immunotherapy)
Show Figures

Graphical abstract

36 pages, 1483 KiB  
Review
Microbial Crosstalk with Therapy: Pharmacomicrobiomics in AML—One Step Closer to Personalized Medicine
by Aneta Nowicka, Hanna Tomczak, Edyta Szałek, Agnieszka Karbownik and Lidia Gil
Biomedicines 2025, 13(7), 1761; https://doi.org/10.3390/biomedicines13071761 - 18 Jul 2025
Viewed by 600
Abstract
Increasing evidence demonstrates the mutualistic connection between the microbiome and acute myeloid leukemia (AML) treatment. Drugs disrupt the microbial balance and, conversely, changes in the microbiome influence therapy. A new field, pharmacomicrobiomics, examines the role of the microbiome in pharmacokinetics, pharmacodynamics, and drug [...] Read more.
Increasing evidence demonstrates the mutualistic connection between the microbiome and acute myeloid leukemia (AML) treatment. Drugs disrupt the microbial balance and, conversely, changes in the microbiome influence therapy. A new field, pharmacomicrobiomics, examines the role of the microbiome in pharmacokinetics, pharmacodynamics, and drug toxicity. The multimodal therapeutic management of AML, along with disease-related immunosuppression, infection, and malnutrition, creates the unique microbial profile of AML patients, in which every delicate modification plays a crucial role in pharmacotherapy. While both preclinical and real-world data have confirmed a bilateral connection between standard chemotherapy and the microbiome, the impact of novel targeted therapies and immunotherapy remains unknown. Multi-omics technologies have provided qualitative and mechanistic insights into specific compositional and functional microbial signatures associated with the outcomes of AML therapy, but require a large-scale investigation to draw reliable conclusions. In this review, we outline the role of the microbiome within the therapeutic landscape of AML, focusing on the determinants of post-treatment dysbiosis and its effects on the therapeutic response and toxicity. We explore emerging strategies for microbiota modulation, highlighting their safety and efficacy. Advances in microbiome-based approaches are an inevitable step toward precision medicine in AML. However, clinical research in a well-defined group of immunocompromised patients is needed to study their variable effects on human health and determine safety issues. Full article
(This article belongs to the Collection Feature Papers in Microbiology in Human Health and Disease)
Show Figures

Figure 1

25 pages, 697 KiB  
Systematic Review
Comparative Meta-Analysis of Survival, Risk, and Treatment Efficacy in Immunotherapy for Metastatic Melanoma Using Random-, Fixed-, and Mixed-Effects Models
by Jelena Ivetić, Jovana Dedeić, Srđan Milićević, Katarina Vidojević and Marija Delić
J. Clin. Med. 2025, 14(14), 5017; https://doi.org/10.3390/jcm14145017 - 15 Jul 2025
Viewed by 318
Abstract
Background: Immune checkpoint inhibitors (ICIs) have reshaped the treatment landscape of metastatic melanoma. While combination regimens often demonstrate improved response and survival compared to monotherapy, they are also associated with a higher incidence of immune-related adverse events (irAEs). Understanding the balance between benefit [...] Read more.
Background: Immune checkpoint inhibitors (ICIs) have reshaped the treatment landscape of metastatic melanoma. While combination regimens often demonstrate improved response and survival compared to monotherapy, they are also associated with a higher incidence of immune-related adverse events (irAEs). Understanding the balance between benefit and risk is essential for making informed treatment decisions, especially given the variability in reported outcomes across clinical trials. Methods: We conducted a systematic review and meta-analysis of 14 clinical trials (comprising 22 treatment arms and >5000 patients) comparing ICI monotherapy (nivolumab, ipilimumab, or pembrolizumab) and combination therapy (nivolumab + ipilimumab) in advanced melanoma. Treatment-related outcomes were synthesized using fixed-effects, random-effects, or generalized linear mixed models (GLMMs), depending on study variability. Survival data were extracted from published Kaplan–Meier curves and analyzed using longitudinal GLMMs to capture trends over time. Results: Compared to monotherapy, combination immunotherapy achieved higher clinical benefit, with an overall response of 52.2% (vs. 31.6%), a five-year overall survival of 55.7% (vs. 34.3%), and a five-year progression-free survival of 39.0% (vs. 17.2%). However, this benefit came with a higher risk of toxicity: immune-related adverse events occurred in 93.2% of patients receiving combination therapy versus in 81.9% receiving monotherapy. Differences were consistent across all individual severe toxicities. Conclusions: Combination immunotherapy offers greater long-term clinical benefit than monotherapy in metastatic melanoma but at the cost of increased toxicity. By applying models adapted to study variability, we provide more reliable estimates of treatment efficacy and risk. GLMMs provide the most robust estimates and enable the modeling of survival dynamics over time. These findings support evidence-based decision-making and highlight the value of model-informed meta-analysis in oncology. Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Treatment of Skin Cancer)
Show Figures

Figure 1

26 pages, 1735 KiB  
Perspective
Optimizing Adjuvant Care in Early Breast Cancer: Multidisciplinary Strategies and Innovative Models from Canadian Centers
by Angela Chan, Nancy Nixon, Muna Al-Khaifi, Alain Bestavros, Christine Blyth, Winson Y. Cheung, Caroline Hamm, Thomas Joly-Mischlich, Mita Manna, Tom McFarlane, Laura V. Minard, Sarah Naujokaitis, Christine Peragine, Cindy Railton and Scott Edwards
Curr. Oncol. 2025, 32(7), 402; https://doi.org/10.3390/curroncol32070402 - 14 Jul 2025
Viewed by 647
Abstract
The adjuvant treatment landscape for hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2–) early breast cancer (EBC) is rapidly evolving, with a diverse range of therapeutic options—including endocrine therapies, bisphosphonates, ovarian function suppression, olaparib, CDK4/6 inhibitors, and emerging agents such as [...] Read more.
The adjuvant treatment landscape for hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2–) early breast cancer (EBC) is rapidly evolving, with a diverse range of therapeutic options—including endocrine therapies, bisphosphonates, ovarian function suppression, olaparib, CDK4/6 inhibitors, and emerging agents such as immunotherapy. While these advances have markedly improved patient outcomes, they also introduce challenges related to implementation, monitoring, and resource allocation. Notably, therapies like CDK4/6 inhibitors require particularly close monitoring, creating logistical and capacity challenges for medical oncologists, whose workloads are already stretched due to rising cancer incidence and treatment complexities. These challenges underscore the need for innovative care delivery solutions to ensure patients with EBC continue to receive optimal care. This paper offers a comprehensive guide—a playbook—of multidisciplinary-team-based care models designed to optimize adjuvant treatment delivery in EBC. Drawing on real-world evidence and successful applications across Canadian centers, we explore models led by nurses, nurse practitioners (NPs), general practitioners in oncology (GPO), and pharmacists. Each model leverages the unique expertise of its team to manage treatment toxicities, facilitate adherence, and enhance patient education, thereby promoting effective and sustainable care delivery. Importantly, these models are not intended to compete with one another, but rather to serve as a flexible recipe book from which breast cancer care teams can draw strategies tailored to their local resources and patient needs. By detailing implementation strategies, benefits, and challenges—in many instances supported by quantitative metrics and economic evaluations—this work aims to inspire care teams nationwide to optimize the adjuvant management of patients with HR+, HER2– EBC. Full article
(This article belongs to the Section Breast Cancer)
Show Figures

Figure 1

15 pages, 452 KiB  
Systematic Review
The Efficacy of Neoantigen-Loaded Dendritic Cell Vaccine Immunotherapy in Non-Metastatic Gastric Cancer
by Menelaos Papakonstantinou, Paraskevi Chatzikomnitsa, Areti Danai Gkaitatzi, Athanasia Myriskou, Alexandros Giakoustidis, Dimitrios Giakoustidis and Vasileios N. Papadopoulos
Med. Sci. 2025, 13(3), 90; https://doi.org/10.3390/medsci13030090 - 11 Jul 2025
Viewed by 992
Abstract
Introduction: Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide. Even though surgery and chemotherapy are the mainstay of treatment, immunotherapy, and more specifically anti-tumor vaccination, has gained popularity over the past years due to the lower related toxicity and [...] Read more.
Introduction: Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide. Even though surgery and chemotherapy are the mainstay of treatment, immunotherapy, and more specifically anti-tumor vaccination, has gained popularity over the past years due to the lower related toxicity and fewer long-term side effects. Dendritic cell (DC) vaccines have been shown to induce tumor specific cytotoxic T-cell (CTL) responses both in vitro and in vivo; however, due to the nature of the disease, resistance to immunotherapy is often developed. Various modifications, such as the implementation of viral vectors, tumor RNA, or even tumor-specific peptides (neoantigens), have been studied as a means to avoid resistance and enhance the effectiveness of the vaccines. In this review, we aim to assess the effects of neoantigen-loaded DC vaccines (naDCVs) on the immune response against gastric cancer cells. Materials and methods: A thorough literature search was conducted on PubMed and clinicaltrials.gov for studies assessing the efficacy of naDCVs against gastric cancer both in vivo and in vitro. The studies were assessed for eligibility by two independent reviewers based on predetermined inclusion and exclusion criteria. The search was completed following the PRISMA guidelines. Results: Eleven studies were included in our systematic review. In five of the studies, the effects of the naDCVs were tested in vitro; in two and in four they were examined both in vitro and in vivo. The in vitro studies showed that the naDCVs resulted in a more robust immune response against the cancer cells in the study groups compared to the control groups. The in vivo studies conducted on mice showed that tumor volume was reduced in the groups treated with the naDCV compared to the untreated groups. What is more, the cytotoxic effect of CTLs against tumor cells was also increased in the vaccine groups. One of the studies was conducted on humans as a phase I study. The results show increased CTL proliferation and cytokine production in the vaccinated group compared to the control, but no difference regarding the tumor size was observed. Conclusions: Neoantigen-loaded DC vaccines can stimulate a strong immune response against specific gastric cancer cell peptides and enhance tumor cell lysis, therefore hindering or even reversing disease progression, offering great potential for the treatment of patients with gastric cancer. Full article
(This article belongs to the Special Issue Feature Papers in Section Cancer and Cancer-Related Diseases)
Show Figures

Figure 1

17 pages, 1548 KiB  
Article
CD19-ReTARGTPR: A Novel Fusion Protein for Physiological Engagement of Anti-CMV Cytotoxic T Cells Against CD19-Expressing Malignancies
by Anne Paulien van Wijngaarden, Isabel Britsch, Matthias Peipp, Douwe Freerk Samplonius and Wijnand Helfrich
Cancers 2025, 17(14), 2300; https://doi.org/10.3390/cancers17142300 - 10 Jul 2025
Viewed by 411
Abstract
Background/Objectives: The physiological activation of cytotoxic CD8pos T cells (CTLs) relies on the engagement of the TCR/CD3 complex with cognate peptide-HLA class I (pHLA-I) on target cells, triggering cell lysis with appropriate cytokine release and minimized off-target toxicity. In contrast, current [...] Read more.
Background/Objectives: The physiological activation of cytotoxic CD8pos T cells (CTLs) relies on the engagement of the TCR/CD3 complex with cognate peptide-HLA class I (pHLA-I) on target cells, triggering cell lysis with appropriate cytokine release and minimized off-target toxicity. In contrast, current immunotherapies for CD19-expressing hematological malignancies, such as chimeric antigen receptor (CAR) T cells and bispecific T cell engagers (BiTEs), bypass TCR/pHLA interactions, resulting in CTL hyperactivation and excessive cytokine release, which frequently cause severe immune-related adverse events (irAEs). Thus, there is a pressing need for T cell-based therapies that preserve physiological activation while maintaining antitumor efficacy. Methods: To address this, we developed CD19-ReTARGTPR, a novel fusion protein consisting of the immunodominant cytomegalovirus (CMV) pp65-derived peptide TPRVTGGAM (TPR) covalently presented by a soluble HLA-B*07:02/β2-microglobulin complex fused to a high-affinity CD19-targeting Fab antibody fragment. The treatment of CD19-expressing cancer cells with CD19-ReTARGTPR makes them recognizable for pre-existing anti-CMVpp65 CTLs via physiological TCR-pHLA engagement. Results: Our preclinical data demonstrate that CD19-ReTARGTPR efficiently redirects anti-CMV CTLs to eliminate CD19-expressing cancer cells, including both established cell lines and primary chronic lymphocytic leukemia (CLL) cells. Unlike CD19-directed CAR T cells or the CD19/CD3 BiTE blinatumomab, CD19-ReTARGTPR mediated robust cytotoxic activity without triggering supraphysiological cytokine release. Importantly, this approach retained efficacy even against cancer cells with low CD19 expression. Conclusions: In summary, we provide a robust proof-of-concept study and show that CD19-ReTARGTPR offers a promising alternative strategy for T cell redirection, enabling the selective and effective killing of CD19-expressing malignancies while minimizing cytokine-driven toxicities through physiological CTL activation pathways. Full article
(This article belongs to the Special Issue New Insights of Hematology in Cancer)
Show Figures

Graphical abstract

9 pages, 517 KiB  
Perspective
Cancer Immunotherapy: The Role of Nursing in Patient Education, Assessment, Monitoring, and Support
by Parmis Mirzadeh, Edith Pituskin, Ivan Au, Sheri Sneath and Catriona J. Buick
Curr. Oncol. 2025, 32(7), 392; https://doi.org/10.3390/curroncol32070392 - 9 Jul 2025
Viewed by 489
Abstract
The prevalence of cancer is rising both in Canada and across the world, with approximately 35 million new cases predicted by 2050. Cancer immunotherapy is a form of treatment that harnesses the body’s immune system to fight cancer cells, increasing life expectancy beyond [...] Read more.
The prevalence of cancer is rising both in Canada and across the world, with approximately 35 million new cases predicted by 2050. Cancer immunotherapy is a form of treatment that harnesses the body’s immune system to fight cancer cells, increasing life expectancy beyond what traditional treatments offer. Immunotherapy may cause immune-related adverse events that differ from the toxicities of traditional treatments. While these events can be detrimental to health, it is critical that they are caught early. This perspective paper examines the evolving role of oncology nurses within the cancer care continuum in caring for patients receiving cancer immunotherapy, specifically immune checkpoint inhibitors. Oncology nurses provide care in many areas, specifically in educating patients on the early detection of side effects to prevent negative outcomes, assessing and monitoring patient symptoms through a variety of means, including nurse-led clinics, and providing support to patients undergoing therapy. This work helps identify gaps in the literature. Future research is required for advancing cancer immunotherapies and better detecting early signs of side effects for nurses practicing in different settings, ensuring timely care. Full article
(This article belongs to the Special Issue Feature Reviews in Section "Oncology Nursing")
Show Figures

Figure 1

37 pages, 1448 KiB  
Review
Immuno-Oncology at the Crossroads: Confronting Challenges in the Quest for Effective Cancer Therapies
by Claudiu Natanael Roznovan, Luminița Gabriela Măruțescu and Gratiela Gradisteanu Pircalabioru
Int. J. Mol. Sci. 2025, 26(13), 6177; https://doi.org/10.3390/ijms26136177 - 26 Jun 2025
Viewed by 710
Abstract
Immuno-oncology has rapidly evolved into a cornerstone of modern cancer therapy, offering promising avenues for durable responses and personalized treatment strategies. This narrative review provides a thorough overview of the mechanisms underlying tumor–immune system interactions and the therapeutic innovations emerging from this knowledge. [...] Read more.
Immuno-oncology has rapidly evolved into a cornerstone of modern cancer therapy, offering promising avenues for durable responses and personalized treatment strategies. This narrative review provides a thorough overview of the mechanisms underlying tumor–immune system interactions and the therapeutic innovations emerging from this knowledge. Central to this discussion is the tumor microenvironment (TME), a complex ecosystem of immune and stromal cells that supports tumor growth and shapes therapeutic outcomes. Key cellular and molecular factors within the TME are examined, along with diverse immune escape strategies. We further analyze the landscape of immunotherapeutic approaches, including immune checkpoint inhibitors, cancer vaccines, adoptive cell therapies such as CAR-T cells, and cytokine-based interventions. This review also addresses the increasing importance of predictive biomarkers in immuno-oncology, particularly in patient stratification, monitoring resistance, and managing immunotherapy-related toxicity. Finally, we explore the emerging role of the microbiome as a modulator of immunotherapy efficacy, shedding light on host–microbe–immune interactions that may influence clinical outcomes. By integrating current biological insights with therapeutic innovation, this review outlines the challenges and opportunities ahead in immuno-oncology and emphasizes the need for translational research and cross-disciplinary collaboration to optimize cancer immunotherapy in the era of precision medicine. Full article
(This article belongs to the Special Issue Biomarkers in Cancer Immunology)
Show Figures

Figure 1

31 pages, 7673 KiB  
Review
Phospholipase A2—A Significant Bio-Active Molecule in Honeybee (Apis mellifera L.) Venom
by Mara Muntean and Adrian Florea
Molecules 2025, 30(12), 2623; https://doi.org/10.3390/molecules30122623 - 17 Jun 2025
Viewed by 584
Abstract
Phospholipase A2 (PLA2) is a prevalent molecule in the honeybee venom. Its importance is reflected by the number of scientists focused on studying it from various points of view. This review summarises a significant amount of data concerning this fascinating [...] Read more.
Phospholipase A2 (PLA2) is a prevalent molecule in the honeybee venom. Its importance is reflected by the number of scientists focused on studying it from various points of view. This review summarises a significant amount of data concerning this fascinating substance. Firstly, the origin and occurrence of PLA2, with similarities and differences among species or populations of bees are highlighted. Next, its synthesis, post-translational processing and structural features are described, followed by the PLA2 availability. In a larger section, the multiple effects of honeybee venom PLA2 are detailed, starting with the main ability as an enzyme to interact with biological membranes and to hydrolyse the sn-2 ester bond in 1,2-diacyl-sn-3-phosphoglycerides; the docking process, the substrate binding and the catalytic steps are analysed too. Then, the pro-/anti-inflammatory effect and allergenic property, the anticoagulant effect and the involvement of PLA2 in apoptosis are revised. Selected antiviral, antibiotic and antitumoral effects of PLA2, as well as its use in immunotherapy are mentioned as beneficial applications. Additionally, the mechanisms of toxicity of PLA2 are presented in detail. Finally, a number of anti-PLA2 compounds are enumerated. In each section, the features of the honeybee venom molecule are discussed in relation to PLA2s from other species. Full article
Show Figures

Figure 1

18 pages, 2608 KiB  
Article
Immune-Related Cutaneous Adverse Events Display Distinct Clinical and Molecular Characteristics, Depending on Immune Checkpoints Targeted
by Lukas Kraehenbuehl, Nicola Winkelbeiner, Patrick Turko, Ramon Staeger, Adhideb Ghosh, Vivienn Kaiser, Pia-Charlotte Stadler, Thierry M. Nordmann, Marie-Charlotte Brüggen, Mitchell P. Levesque, Emmanuel Contassot, Lars E. French, Reinhard Dummer and Barbara Meier-Schiesser
Cancers 2025, 17(12), 1992; https://doi.org/10.3390/cancers17121992 - 14 Jun 2025
Viewed by 661
Abstract
Background/Objectives: Immune-related cutaneous adverse events (ircAEs) are common complications of cancer immunotherapy and provide insight into immune-related adverse events (irAEs) more broadly. To enhance our molecular understanding, we characterized ircAEs resulting from single-agent (PD1) and combined immunotherapy regimens (P+C). Clinically, maculopapular rash [...] Read more.
Background/Objectives: Immune-related cutaneous adverse events (ircAEs) are common complications of cancer immunotherapy and provide insight into immune-related adverse events (irAEs) more broadly. To enhance our molecular understanding, we characterized ircAEs resulting from single-agent (PD1) and combined immunotherapy regimens (P+C). Clinically, maculopapular rash (MPR) and toxic epidermal necrolysis (TEN) resemble ircAEs, providing a valuable basis for investigations. Methods: To investigate the transcriptome and immune infiltrates in ircAEs, we conducted transcriptomic analyses and multiplexed immunohistochemistry on skin biopsies from patients receiving PD1 and P+C, as well as those with MPR, TEN, and healthy controls. Results: Principal component analysis revealed distinct transcriptomic clustering between ircAEs, MPR, and TEN. Specifically, PD1 ircAEs exhibited a gene expression profile similar to TEN, with upregulation of Type-I-response-related genes (e.g., CXCL9 Log2FC 5.34, p < 0.0001, CXCL10 Log2FC 6.03, p < 0.0001), while P+C ircAEs more closely resembled MPR. Immune infiltrates differed significantly between all groups (p = 0.002 by PERMANOVA for all groups). CD4 T-cells were abundant in the dermis of ircAEs from any type of immunotherapy. However, PD1 stained positive in 1.07% of CD4 cells with PD1 monotherapy, compared to 0.3%, 0.4%, and 0.08% in P+C, MPR, and TEN, respectively. Conclusions: This study identified distinct molecular and cellular signatures in ircAEs depending on the type of immune checkpoint blockade. aPD1-associated ircAEs share similarities with the cytotoxic profile of TEN, while P+C more closely mirrored MPR. These findings support the need for tailored management strategies for ircAEs, emphasizing personalized therapeutic approaches to minimize treatment interruptions while preserving the efficacy of cancer immunotherapy. Full article
(This article belongs to the Special Issue Novel Developments on Skin Cancer Diagnostics and Treatment)
Show Figures

Figure 1

18 pages, 2085 KiB  
Article
PET Radiomics Signatures and Artificial Intelligence for Decoding Immunotherapy Response in Advanced Cutaneous Squamous Cell Carcinoma: A Retrospective Single-Center Study
by Luigi Manco, Ilaria Proietti, Giovanni Scribano, Riccardo Pirisino, Oreste Bagni, Concetta Potenza, Giovanni Pellacani and Luca Filippi
Appl. Sci. 2025, 15(12), 6453; https://doi.org/10.3390/app15126453 - 8 Jun 2025
Viewed by 533
Abstract
The aim of this study was to develop a baseline [18F]FDG PET/CT model to predict immunotherapy response in advanced cutaneous squamous cell carcinoma (cSCC) and noninvasively determine tumor grade, thereby enhancing early patient stratification. We retrospectively analyzed 59 patients with histologically [...] Read more.
The aim of this study was to develop a baseline [18F]FDG PET/CT model to predict immunotherapy response in advanced cutaneous squamous cell carcinoma (cSCC) and noninvasively determine tumor grade, thereby enhancing early patient stratification. We retrospectively analyzed 59 patients with histologically confirmed advanced cSCC submitted to immunotherapy with cemiplimab. All underwent [18F]FDG PET/CT at baseline and after approximately 12 weeks. Clinical response was assessed through PET findings integrated with clinical and dermatological evaluation, and patients were classified as responders (complete/partial metabolic response or stable disease) or non-responders (progression or toxicity-related discontinuation). Tumors were also classified as low to intermediate (G1–G2) or poorly differentiated (G3). Machine learning models (Random Forest and Extreme Gradient Boosting) were trained to predict treatment response and tumor grade. Clinical benefit was observed in 46/59 patients (77.9%), while 13 (22.1%) were non-responders. Histology showed 64.4% (n = 38) G1–G2 and 35.6% (n = 21) G3 tumors. The PET-based model best predicted clinical benefit (AUC = 0.96, accuracy = 91% cross-validation; AUC = 0.88, accuracy = 82% internal validation). For tumor grade prediction, the CT-based model achieved a higher AUC of 0.80 (accuracy 73%), whereas the PET-based model reached an AUC of 0.78 but demonstrated a slightly higher accuracy of 77%. Radiomic analysis of baseline [18F]FDG PET enables the discriminative prediction of immunotherapy response and tumor grade in advanced cSCC, with PET-based models outperforming CT-based ones. Full article
Show Figures

Figure 1

23 pages, 1878 KiB  
Article
Quality of Life of Lung Cancer Patients with Immune-Related Endocrinopathies During Immunotherapy: A Prospective Study Based on the EORTC QLQ-C30 and QLQ-LC13 Questionnaires in Romania
by Simona Coniac, Mariana Cristina Costache-Outas, Ionuţ-Lucian Antone-Iordache, Alexandra-Valentina Anghel, Maria-Alexandra Bobolocu, Andreea Zamfir, Horia-Dan Liscu, Andreea-Iuliana Ionescu and Corin Badiu
Curr. Oncol. 2025, 32(6), 332; https://doi.org/10.3390/curroncol32060332 - 5 Jun 2025
Viewed by 1520
Abstract
(1) Background: Globally, lung cancer is the leading cause of cancer death, but immunotherapy has impressively improved the outcomes, generating longer progression-free survival and overall survival. Endocrine immune-related adverse events (irAEs) are mostly irreversible and need life-long hormonal substitution therapy. The evaluation of [...] Read more.
(1) Background: Globally, lung cancer is the leading cause of cancer death, but immunotherapy has impressively improved the outcomes, generating longer progression-free survival and overall survival. Endocrine immune-related adverse events (irAEs) are mostly irreversible and need life-long hormonal substitution therapy. The evaluation of the quality of life of lung cancer patients experiencing endocrine toxicities during immune checkpoint inhibitor (ICI) treatment is relevant for both patients and healthcare providers. (2) Methods: This was a prospective cohort study of lung cancer patients treated with immune checkpoint inhibitors in a tertiary-level hospital in Romania from 1 December 2021 to 30 September 2024. Quality of life was assessed using versions of the EORTC QLQ-C30 and EORTC QLQ-LC-13 validated and translated into the Romanian language. We investigated several clinical variables to evaluate their impact on QoL. (3) Results: Fifty-nine lung cancer patients were evaluated for their QoL before ICI initiation and at the end of the study. Endocrine-irAEs occurred in 17 lung cancer patients (28.8%). Quality of life as assessed using the EORTC questionnaires was statistically significantly improved, even in patients experiencing endocrine-irAEs. (4) Conclusions: Our prospective cohort study succeeded in delivering the proof of concept of an increased QoL in lung cancer patients who had developed endocrine-irAEs under immunotherapy. Despite toxicities, especially rather frequent endocrine-irAEs, ICIs enabled durable disease control and symptom relief, improving the QoL of the overall trial population. As more lung cancer patients undergo immunotherapy in metastatic or early stages, we draw attention to this particular patient population with autoimmune endocrinopathies, as they will live longer and require life-long hormonal therapy. Full article
(This article belongs to the Special Issue Palliative Care and Supportive Medicine in Cancer)
Show Figures

Figure 1

10 pages, 549 KiB  
Article
Complete Blood Count-Derived Biomarkers’ Association with Risk of PD-1 or PD-1/CTLA-4 Inhibitor-Induced Hypothyroidism in Patients with Solid Tumors
by Ketevan Lomidze, Nino Kikodze, Marine Gordeladze, Nino Charkviani and Tinatin Chikovani
Immuno 2025, 5(2), 21; https://doi.org/10.3390/immuno5020021 - 4 Jun 2025
Viewed by 561
Abstract
Background: A novel and highly effective strategy for tumor immunotherapy involves enhancing host immune responses against tumors through the blockade of checkpoint molecules. The most common toxicities associated with checkpoint blockade therapies include autoimmune damage to various organs. Purpose: This study aims to [...] Read more.
Background: A novel and highly effective strategy for tumor immunotherapy involves enhancing host immune responses against tumors through the blockade of checkpoint molecules. The most common toxicities associated with checkpoint blockade therapies include autoimmune damage to various organs. Purpose: This study aims to investigate hematological markers derived from complete blood counts (CBCs)—including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), derived neutrophil-to-lymphocyte ratio (dNLR), white blood cell-to-hemoglobin ratio (WHR), neutrophils, lymphocytes, platelets, hemoglobin, red blood cell (RBC) count, neutrophil-to-RBC ratio (NRR), and neutrophil-to-hemoglobin ratio (NHR)—as potential prognostic biomarkers for the early identification of hypothyroidism in patients receiving PD-1 or PD-1/CTLA-4 immune checkpoint inhibitors. Materials and Methods: A prospective observational study was conducted on 44 patients with stage III-IV solid tumors treated with immune checkpoint (PD-1 or PD-1/CTLA-4) inhibitors. Thyroid function tests and CBC-derived biomarkers were collected at baseline, before immunotherapy. In the immunotherapy cohort, 15 of the 44 patients developed immune-related hypothyroidism, defined as overt autoimmune thyroiditis (TSH > 4.0, FT4 < 12, and anti-TPO antibodies > 30 IU/mL and/or anti-TG antibodies > 95 IU/mL) (Group 1). In comparison, 29 patients maintained normal thyroid function (Group 2). The control group comprised 14 age- and sex-matched healthy volunteers (Group 3). Statistical analyses were performed using analysis of variance (ANOVA) to compare blood parameters among the three groups (Group 1, Group 2, and Group 3) before treatment, with statistical significance set at a p-value < 0.05. Receiver operating characteristic (ROC) curve analysis was conducted to evaluate the diagnostic power of the potential prognostic biomarkers areas. The area under the curve (AUC), sensitivity, and specificity were calculated for the 44 immunotherapy patients. Results: The PLR was significantly higher (262.25 ± 162.95), while WBCs-neutrophils, the WHR, the NRR, the NHR, WBCs, neutrophils, and lymphocytes were lower (2.07 ± 0.66, 0.54 ± 0.19, 0.96 ± 0.28, 0.36 ± 0.14, 6.36 ± 2.07, 4.29 ± 1.55, and 1.23 ± 0.41, respectively) at baseline in Group 1 in comparison to Group 2. ROC curve analysis revealed that the areas under the curve (AUC) for WBCs, neutrophils, lymphocytes, WBCs-neutrophils, the PLR, the WHR, the NRR, and the NHR were 0.9, 0.87, 0.83, 0.85, 0.84, 0.92, 0.89, and 0.87, respectively. These values exceeded the threshold, indicating the high prognostic potential of each marker. Conclusions: Lower baseline levels of WBCs-neutrophils, the WHR, the NRR, the NHR, WBCs, neutrophils, and lymphocytes, along with a higher PLR, were associated with an increased risk of hypothyroidism in patients receiving PD-1 or PD-1/CTLA-4 inhibitors. These CBC-derived biomarkers represent simple, accessible, and potentially useful tools for predicting hypothyroidism in cancer patients undergoing immunotherapy. Further studies in bigger cohorts are needed to validate our findings. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

Back to TopTop