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

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

Search Results (9,513)

Search Parameters:
Keywords = immunotherapies

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 1130 KiB  
Systematic Review
Allergy to Plant-Based Panallergens LTPs in Children: A Scoping Review
by Nikos Priftis, Dimitra Karaviti and Kostas Douros
Allergies 2024, 4(4), 218-233; https://doi.org/10.3390/allergies4040015 (registering DOI) - 18 Nov 2024
Viewed by 36
Abstract
Introduction: Lipid Transfer Proteins (LTPs) are plant-derived panallergens that have emerged as significant allergens in Mediterranean populations. Though less common in children, LTP allergies represent a critical consideration for physicians diagnosing plant food allergies in this demographic. Methodology: PRISMA-ScR guidelines were followed. A [...] Read more.
Introduction: Lipid Transfer Proteins (LTPs) are plant-derived panallergens that have emerged as significant allergens in Mediterranean populations. Though less common in children, LTP allergies represent a critical consideration for physicians diagnosing plant food allergies in this demographic. Methodology: PRISMA-ScR guidelines were followed. A search with specific terms was performed in searchable databases. Two of the authors extracted and evaluated the data. Results: A total of 21 original studies and 6 case reports focusing on LTP allergies in the paediatric population met the inclusion criteria. Diagnostic tools, predictive markers and management options for LTP allergies were examined. Allergens, clinical presentation and history were the diagnostic tools investigated. The clinical and laboratory phenotypes of the patient were considered possible predictive markers for the evaluation and progression of LTP allergies. Lastly, dietary modifications and sublingual immunotherapy were identified as the main focus of LTP allergy management. Discussion: A summary of the results is presented, and at the same time, questions concerning the nature of LTP allergies and their management are raised. Conclusions: LTP allergy in children is something physicians should be aware of. Further research is needed to establish the differences in LTP allergies in children and adults and the effectiveness of immunotherapy in paediatric populations. Full article
(This article belongs to the Section Allergen/Pollen)
Show Figures

Figure 1

11 pages, 3642 KiB  
Case Report
Inflammatory Mesenteric Disease and Sarcoidosis-like Reaction in a Patient with Lung Adenocarcinoma Who Received Pembrolizumab: Paraneoplastic Syndrome, Secondary to Checkpoint Inhibitor or Chance Finding?
by Luis Posado-Domínguez, María Escribano-Iglesias, Lorena Bellido-Hernández, Johana Gabriela León-Gil, María Asunción Gómez-Muñoz, Felipe Gómez-Caminero López, María Martín-Galache, Sandra M. Inés-Revuelta and Emilio Fonseca-Sánchez
Curr. Oncol. 2024, 31(11), 7319-7329; https://doi.org/10.3390/curroncol31110540 (registering DOI) - 18 Nov 2024
Viewed by 111
Abstract
Summary: Anti PD1/PD-L1 agents, including pembrolizumab, have revolutionized the oncological treatment of different types of cancer, including non-small cell lung cancer. The most frequent complications associated with this type of treatment are mild and are located at the thyroid, pulmonary or hepatic [...] Read more.
Summary: Anti PD1/PD-L1 agents, including pembrolizumab, have revolutionized the oncological treatment of different types of cancer, including non-small cell lung cancer. The most frequent complications associated with this type of treatment are mild and are located at the thyroid, pulmonary or hepatic level. Sarcoid like reaction and mesenteric panniculitis secondary to pembrolizumab treatment are two very rare adverse effects. We present the case of a patient with these complications. Purpose: the treatment of metastatic non-small cell lung cancer has undergone a major change in the last 10 years, largely due to the advent of immunotherapy. Anti PD1 agents such as pembrolizumab have increased the median survival of these patients from 13 to 26 months. Most frequent immunorelated side effects are hypothyroidism, pneumonitis or elevated liver enzymes. However, there are other adverse effects, including sarcoid-like reaction and mesenteric panniculitis, which should be known by the professionals involved in the diagnosis and treatment of this type of patient. We present the case of a 62-year-old man with a history of unresectable and non-irradiable stage IIIB epidermoid lung carcinoma with a PD-L1 expression of 30% in whom pembrolizumab was discontinued after 4 cycles due to immunorelated arthritis. One year later he consulted for severe abdominal pain. A PET-CT scan was performed, showing hilar lymphadenopathy and inflammation of abdominal mesenteric fat. A biopsy of lesions in both areas showed non-necrotizing granulomatous lymphadenitis in hilar adenopathy and patchy fibrosis of mesenteric fat. The picture was classified as sarcoidosis-like reaction and mesenteric panniculitis secondary to pembrolizumab. Anti-PD1 agents cause hyperactivation of the immune system through T-cell proliferation. Sarcoid-like reaction is a very rare complication that can mask progressive tumor disease. Awareness of immunorelated complications by oncologists, internists, and radiologists is important for an appropriate diagnostic approach and targeted test ordering. Full article
(This article belongs to the Section Thoracic Oncology)
Show Figures

Figure 1

12 pages, 592 KiB  
Review
Exploration of the Dual Role of Dectin-1 in Tumor Development and Its Therapeutic Potential
by Yuxuan Cai and Ke Wu
Curr. Oncol. 2024, 31(11), 7275-7286; https://doi.org/10.3390/curroncol31110536 (registering DOI) - 17 Nov 2024
Viewed by 190
Abstract
Immunotherapy, particularly immune checkpoint inhibitors like PD-1, PD-L1, and CTLA-4, has revolutionized cancer treatment. However, the role of the innate immune system, especially pattern recognition receptors, in cancer development and immunity is gaining more and more attention. Dectin-1, a C-type lectin receptor primarily [...] Read more.
Immunotherapy, particularly immune checkpoint inhibitors like PD-1, PD-L1, and CTLA-4, has revolutionized cancer treatment. However, the role of the innate immune system, especially pattern recognition receptors, in cancer development and immunity is gaining more and more attention. Dectin-1, a C-type lectin receptor primarily involved in antifungal immunity, has emerged as a significant player in cancer biology, exhibiting both pro-tumor and anti-tumor roles. This dual function largely depends on the tumor type and microenvironment. Dectin-1 can promote immune responses against tumors like melanoma and breast cancer by enhancing both innate and adaptive immunity. However, in tumors like pancreatic ductal adenocarcinoma and colorectal cancer, Dectin-1 activation suppresses T cell immunity, facilitating tumor progression. This review explores the complex mechanisms by which Dectin-1 modulates the tumor microenvironment and discusses its potential as a therapeutic target for cancer treatment. Full article
Show Figures

Figure 1

22 pages, 3627 KiB  
Article
Can the Analysis of Toll-like Receptors (TLR) on NK and NKT-like Cells Improve Gastric Cancer Diagnostics and Treatment?
by Marek Kos, Krzysztof Bojarski, Paulina Mertowska, Sebastian Mertowski, Piotr Tomaka, Monika Zaborek-Łyczba, Jakub Łyczba, Łukasz Dziki and Ewelina Grywalska
Cancers 2024, 16(22), 3854; https://doi.org/10.3390/cancers16223854 (registering DOI) - 17 Nov 2024
Viewed by 374
Abstract
Background/Objectives: The aim of this study was to determine the assessment of the percentage of NK and NKT-like cells expressing Toll-like receptors (TLR-2, TLR-3, TLR-4, and TLR-9) in patients with gastric cancer (GC) compared with healthy volunteers (HV) and to investigate differences [...] Read more.
Background/Objectives: The aim of this study was to determine the assessment of the percentage of NK and NKT-like cells expressing Toll-like receptors (TLR-2, TLR-3, TLR-4, and TLR-9) in patients with gastric cancer (GC) compared with healthy volunteers (HV) and to investigate differences according to cancer subtype. We also assessed TLR gene expression by RT-qPCR to assess whether TLRs could be diagnostic and prognostic biomarkers. Methods: The study included 86 patients with histologically confirmed gastric cancer and 30 healthy volunteers. Peripheral blood samples were collected from the participants, and TLR expression on NK and NKT-like cells was assessed by flow cytometry and RT-qPCR. The expression of TLR2, TLR3, TLR4, and TLR9 genes was assessed using genetic material derived from NK and NKT-like cells sourced from PBMC. The obtained results were statistically analyzed using Mann–Whitney U and Kruskal–Wallis tests, and the predictive ability of variables was assessed using ROC curve analysis. Results: A significantly higher expression of TLR receptors (TLR-2, TLR-3, TLR-4, and TLR-9) was found in patients with gastric cancer compared to healthy volunteers (p < 0.05). TLR expression also differed depending on the cancer subtype, and higher expression was observed in more advanced GC subtypes. RT-qPCR analysis showed significantly increased expression of TLR genes in the group of GC patients. ROC curves indicate a high ability of TLRs to differentiate between GC patients and healthy individuals. Conclusions: The expression of TLRs on NK and NKT-like cells is clearly increased in patients with gastric cancer, especially in more advanced subtypes of the tumor. The results suggest that TLRs could potentially be used as diagnostic and prognostic biomarkers and represent potential targets for immune therapies in GC. However, further studies are needed to determine the functional role of TLRs in disease progression and the possibility of their use in personalized treatment. Full article
(This article belongs to the Special Issue Molecular Alterations and Targeted Therapy in Gastric Cancer)
Show Figures

Figure 1

12 pages, 3108 KiB  
Article
A Microfluidic-Based Sensing Platform for Rapid Quality Control on Target Cells from Bioreactors
by Alessia Foscarini, Fabio Romano, Valeria Garzarelli, Antonio Turco, Alessandro Paolo Bramanti, Iolena Tarantini, Francesco Ferrara, Paolo Visconti, Giuseppe Gigli and Maria Serena Chiriacò
Sensors 2024, 24(22), 7329; https://doi.org/10.3390/s24227329 (registering DOI) - 16 Nov 2024
Viewed by 334
Abstract
We investigated the design and characterization of a Lab-On-a-Chip (LoC) cell detection system primarily designed to support immunotherapy in cancer treatment. Immunotherapy uses Chimeric Antigen Receptors (CARs) and T Cell Receptors (TCRs) to fight cancer, engineering the response of the immune system. In [...] Read more.
We investigated the design and characterization of a Lab-On-a-Chip (LoC) cell detection system primarily designed to support immunotherapy in cancer treatment. Immunotherapy uses Chimeric Antigen Receptors (CARs) and T Cell Receptors (TCRs) to fight cancer, engineering the response of the immune system. In recent years, it has emerged as a promising strategy for personalized cancer treatment. However, it requires bioreactor-based cell culture expansion and manual quality control (QC) of the modified cells, which is time-consuming, labour-intensive, and prone to errors. The miniaturized LoC device for automated QC demonstrated here is simple, has a low cost, and is reliable. Its final target is to become one of the building blocks of an LoC for immunotherapy, which would take the place of present labs and manual procedures to the benefit of throughput and affordability. The core of the system is a commercial, on-chip-integrated capacitive sensor managed by a microcontroller capable of sensing cells as accurately measured charge variations. The hardware is based on standardized components, which makes it suitable for mass manufacturing. Moreover, unlike in other cell detection solutions, no external AC source is required. The device has been characterized with a cell line model selectively labelled with gold nanoparticles to simulate its future use in bioreactors in which labelling can apply to successfully engineered CAR-T-cells. Experiments were run both in the air—free drop with no microfluidics—and in the channel, where the fluid volume was considerably lower than in the drop. The device showed good sensitivity even with a low number of cells—around 120, compared with the 107 to 108 needed per kilogram of body weight—which is desirable for a good outcome of the expansion process. Since cell detection is needed in several contexts other than immunotherapy, the usefulness of this LoC goes potentially beyond the scope considered here. Full article
(This article belongs to the Section Biosensors)
Show Figures

Figure 1

19 pages, 4184 KiB  
Article
Identification of Functional Immune Biomarkers in Breast Cancer Patients
by Roshanak Derakhshandeh, Yuyi Zhu, Junxin Li, Danubia Hester, Rania Younis, Rima Koka, Laundette P. Jones, Wenji Sun, Olga Goloubeva, Katherine Tkaczuk, Joshua Bates, Jocelyn Reader and Tonya J. Webb
Int. J. Mol. Sci. 2024, 25(22), 12309; https://doi.org/10.3390/ijms252212309 - 16 Nov 2024
Viewed by 156
Abstract
Cancer immunotherapy has emerged as an effective, personalized treatment for certain patients, particularly for those with hematological malignancies. However, its efficacy in breast cancer has been marginal—perhaps due to cold, immune-excluded, or immune-desert tumors. Natural killer T (NKT) cells play a critical role [...] Read more.
Cancer immunotherapy has emerged as an effective, personalized treatment for certain patients, particularly for those with hematological malignancies. However, its efficacy in breast cancer has been marginal—perhaps due to cold, immune-excluded, or immune-desert tumors. Natural killer T (NKT) cells play a critical role in cancer immune surveillance and are reduced in cancer patients. Thus, we hypothesized that NKT cells could serve as a surrogate marker for immune function. In order to assess which breast cancer patients would likely benefit from immune cell-based therapies, we have developed a quantitative method to rapidly assess NKT function using stimulation with artificial antigen presenting cells followed by quantitative real-time PCR for IFN-γ. We observed a significant reduction in the percentage of circulating NKT cells in breast cancer patients, compared to healthy donors; however, the majority of patients had functional NKT cells. When we compared BC patients with highly functional NKT cells, as indicated by high IFN-γ induction, to those with little to no induction, following stimulation of NKT cells, there was no significant difference in NKT cell number between the groups, suggesting functional loss has more impact than physical loss of this subpopulation of T cells. In addition, we assessed the percentage of tumor-infiltrating lymphocytes and PD-L1 expression within the tumor microenvironment in the low and high responders. Further characterization of immune gene signatures in these groups identified a concomitant decrease in the induction of TNFα, LAG3, and LIGHT in the low responders. We next investigated the mechanisms by which breast cancers suppress NKT-mediated anti-tumor immune responses. We found that breast cancers secrete immunosuppressive lipids, and treatment with commonly prescribed medications that modulate lipid metabolism, can reduce tumor growth and restore NKT cell responses. Full article
Show Figures

Graphical abstract

9 pages, 1061 KiB  
Brief Report
Glypican-3 and Cytokeratin-19 Expression in Pancreatic Cancer in a Canadian Population
by Carley Bekkers, Ravi Ramjeesingh and Thomas Arnason
J. Clin. Med. 2024, 13(22), 6893; https://doi.org/10.3390/jcm13226893 (registering DOI) - 16 Nov 2024
Viewed by 281
Abstract
Background/Objectives: One study of pancreatic ductal adenocarcinoma has found expression of glypican-3 (GPC3) and cytokeratin-19 (CK19) determined by immunohistochemistry to be associated with higher stage and grade disease, with a more adverse prognosis. The reported 44% rate of GPC3 expression in pancreatic cancer [...] Read more.
Background/Objectives: One study of pancreatic ductal adenocarcinoma has found expression of glypican-3 (GPC3) and cytokeratin-19 (CK19) determined by immunohistochemistry to be associated with higher stage and grade disease, with a more adverse prognosis. The reported 44% rate of GPC3 expression in pancreatic cancer raises the important possibility that targeted immunotherapies currently in development for hepatocellular carcinoma may also prove useful for GPC3-expressing pancreatic cancers. The present study aims to determine if a similar expression pattern of these markers and stage/grade/prognostic associations is present in our Canadian patient population. Methods: Patients with a pancreatic surgical resection for adenocarcinoma or neuroendocrine tumor (NET) were identified from pathology records over a 5-year period. Immunohistochemistry for GPC3 and CK19 was performed on archived tumor tissue and the proportion of positive cells and intensity of staining were recorded. Grade, stage, and overall survival were compared in patients with NETs that were CK19-positive versus -negative. Results: All 72 pancreatic adenocarcinomas and 20 NETs tested were negative for GPC3, apart from a single case of pancreatic adenocarcinoma. All 72 adenocarcinomas were positive for CK19 expression. Half of the NETs were positive for CK19. There was no correlation between CK19 expression in NETs and tumor grade, lymph node metastasis, distant metastasis, or overall survival. Conclusions: We are skeptical of the reported prognostic value of GPC3 and CK19 in pancreatic adenocarcinomas. CK19 as a prognostic marker in NETs has potential for further study. The results with our protocol for GPC3 immunohistochemistry suggest that pancreatic cancer may be a less promising target for GPC3-targeted immunotherapies than previously thought. Full article
(This article belongs to the Special Issue Pancreatic Cancer: Current and Emerging Treatment Options)
Show Figures

Figure 1

18 pages, 4537 KiB  
Review
Quantitative Approach to Explore Regulatory T Cell Activity in Immuno-Oncology
by Alejandro Serrano, Sara Zalba, Juan Jose Lasarte, Iñaki F. Troconiz, Natalia Riva and Maria J. Garrido
Pharmaceutics 2024, 16(11), 1461; https://doi.org/10.3390/pharmaceutics16111461 - 15 Nov 2024
Viewed by 249
Abstract
The failure of immunotherapies in cancer patients is being widely studied due to the complexities present in the tumor microenvironment (TME), where regulatory T cells (Treg) appear to actively participate in providing an immune escape mechanism for tumors. Therefore, therapies to specifically inhibit [...] Read more.
The failure of immunotherapies in cancer patients is being widely studied due to the complexities present in the tumor microenvironment (TME), where regulatory T cells (Treg) appear to actively participate in providing an immune escape mechanism for tumors. Therefore, therapies to specifically inhibit tumor-infiltrating Treg represent a challenge, because Treg are distributed throughout the body and provide physiological immune homeostasis to prevent autoimmune diseases. Characterization of immunological and functional profiles could help to identify the mechanisms that need to be inhibited or activated to ensure Treg modulation in the tumor. To address this, quantitative in silico approaches based on mechanistic mathematical models integrating multi-scale information from immune and tumor cells and the effect of different therapies have allowed the building of computational frameworks to simulate different hypotheses, some of which have subsequently been experimentally validated. Therefore, this review presents a list of diverse computational mathematical models that examine the role of Treg as a crucial immune resistance mechanism contributing to the failure of immunotherapy. In addition, this review highlights the relevance of certain molecules expressed in Treg that are associated with the TME immunosuppression, which could be incorporated into the mathematical model for a better understanding of the contribution of Treg modulation. Finally, different preclinical and clinical combinations of molecules are also included to show the trend of new therapies targeting Treg. Full article
(This article belongs to the Section Pharmacokinetics and Pharmacodynamics)
Show Figures

Graphical abstract

13 pages, 1417 KiB  
Article
Prognostic Significance of PD-L1 Expression on Circulating Myeloid-Derived Suppressor Cells in NSCLC Patients Treated with Anti-PD-1/PD-L1 Checkpoint Inhibitors
by Roser Salvia, Laura G. Rico, Teresa Morán, Jolene A. Bradford, Michael D. Ward, Ana Drozdowskyj, Joan Climent-Martí, Eva M. Martínez-Cáceres, Rafael Rosell and Jordi Petriz
Int. J. Mol. Sci. 2024, 25(22), 12269; https://doi.org/10.3390/ijms252212269 - 15 Nov 2024
Viewed by 248
Abstract
Even though anti-PD-1/PD-L1 immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) have improved survival, a high percentage of patients still do not respond to ICIs. Myeloid-derived suppressor cells (MDSCs) are circulating cells that express PD-L1 and can infiltrate and proliferate in [...] Read more.
Even though anti-PD-1/PD-L1 immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) have improved survival, a high percentage of patients still do not respond to ICIs. Myeloid-derived suppressor cells (MDSCs) are circulating cells that express PD-L1 and can infiltrate and proliferate in the tumor microenvironment, inducing immunosuppression. By evaluating changes in PD-L1 expression of live peripheral blood MDSCs, we are able to define a new PD-L1 index, useful in predicting ICI escape in NSCLC patients before initiating anti-PD-1/PD-L1 immunotherapy. In this study, a cohort of 37 NSCLC patients was prospectively analyzed, obtaining independent PD-L1 indexes. In patients with a PD-L1 index > 5.88, progressive disease occurred in 58.33% of patients [median progression-free survival (PFS) = 5.73 months; 95%CI = 2.67–20.53], showing significant differences when compared with patients with a PD-L1 index ≤ 5.88, in whom 7.69% progressed and median PFS was not reached (NR); p-value = 0.0042. Overall survival (OS) was significantly worse in patients with a high vs. low PD-L1 index (41.67% vs. 76.92%; median OS = 18.03 months, 95%CI = 6.77–25.23 vs. NR, 95%CI = 1.87-NR; p-value = 0.035). The PD-L1 index can be applied to stratify NSCLC patients according to their probability of response to ICIs at baseline. In addition to quantifying tumoral expression, this index could be used to compare nonresponse to treatment. Full article
(This article belongs to the Special Issue Cancer Biomarker: Current Status and Future Perspectives)
Show Figures

Figure 1

22 pages, 3386 KiB  
Systematic Review
Immunotherapy Responses in Viral Hepatitis-Induced HCC: A Systematic Review and Meta-Analysis
by Junaid Anwar, Hafiz Muhammad Arslan, Zouina Sarfraz, Juwairiya Shuroog, Ahmed Abdelhakeem, Ali Saeed and Anwaar Saeed
Curr. Oncol. 2024, 31(11), 7204-7225; https://doi.org/10.3390/curroncol31110532 - 15 Nov 2024
Viewed by 328
Abstract
Background: Hepatocellular carcinoma (HCC) is a prevalent liver cancer with poor prognosis, often linked to hepatitis B (HBV) and C (HCV) infections. This meta-analysis evaluates the efficacy of immunotherapy in HCC, particularly in cases arising from viral hepatitis. Methods: In adherence [...] Read more.
Background: Hepatocellular carcinoma (HCC) is a prevalent liver cancer with poor prognosis, often linked to hepatitis B (HBV) and C (HCV) infections. This meta-analysis evaluates the efficacy of immunotherapy in HCC, particularly in cases arising from viral hepatitis. Methods: In adherence to PRISMA Statement 2020 guidelines, the immunotherapeutic outcomes comprised objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). Data were analyzed from randomized controlled trials up to April 2024 using the fixed-effects models in R (V.4.3.3.) and RevMan (Cochrane). Results: This study included 9 trials with 5316 patients. The ORR was slightly higher in the viral group at 27.93% compared to 24.07% in the non-viral group, though this difference was not significant (p = 0.15). Viral HCC patients exhibited a median PFS of 7.3 months (IQR: 6.2–8.4) compared to 5.8 months (IQR: 5.48–6.13) in non-viral patients, a significant improvement (p = 0.005). Similarly, median OS was longer in the viral group at 16.8 months (IQR: 12.99–20.61) versus 15.2 months (IQR: 13.25–17.15) for non-viral HCC, which was also significant (p < 0.0001). The median OS for viral HCC was 16.8 months (IQR: 14.11–19.49 months), with HBV patients experiencing slightly higher survival at 17.15 months (IQR: 14.3–20 months) compared to 16.8 months (IQR: 12.99–20.61 months) for HCV patients; this difference was not statistically significant (p = 0.89). Conclusions: Immunotherapy shows potential in treating HCC, with significantly better outcomes in viral HCC, particularly HBV-associated cases. The heterogeneity highlights the need for personalized treatment approaches based on the viral background of HCC patients. Further research should aim to optimize these therapies to improve survival rates. Full article
Show Figures

Figure 1

18 pages, 772 KiB  
Review
Metabolic Reprogramming of Immune Cells in the Tumor Microenvironment
by Jing Wang, Yuanli He, Feiming Hu, Chenchen Hu, Yuanjie Sun, Kun Yang and Shuya Yang
Int. J. Mol. Sci. 2024, 25(22), 12223; https://doi.org/10.3390/ijms252212223 - 14 Nov 2024
Viewed by 391
Abstract
Metabolic reprogramming of immune cells within the tumor microenvironment (TME) plays a pivotal role in shaping tumor progression and responses to therapy. The intricate interplay between tumor cells and immune cells within this ecosystem influences their metabolic landscapes, thereby modulating the immune evasion [...] Read more.
Metabolic reprogramming of immune cells within the tumor microenvironment (TME) plays a pivotal role in shaping tumor progression and responses to therapy. The intricate interplay between tumor cells and immune cells within this ecosystem influences their metabolic landscapes, thereby modulating the immune evasion tactics employed by tumors and the efficacy of immunotherapeutic interventions. This review delves into the metabolic reprogramming that occurs in tumor cells and a spectrum of immune cells, including T cells, macrophages, dendritic cells, and myeloid-derived suppressor cells (MDSCs), within the TME. The metabolic shifts in these cell types span alterations in glucose, lipid, and amino acid metabolism. Such metabolic reconfigurations can profoundly influence immune cell function and the mechanisms by which tumors evade immune surveillance. Gaining a comprehensive understanding of the metabolic reprogramming of immune cells in the TME is essential for devising novel cancer therapeutic strategies. By targeting the metabolic states of immune cells, it is possible to augment their anti-tumor activities, presenting new opportunities for immunotherapeutic approaches. These strategies hold promise for enhancing treatment outcomes and circumventing the emergence of drug resistance. Full article
(This article belongs to the Special Issue Metabolic Pathways in Cancer Cells)
Show Figures

Figure 1

29 pages, 5814 KiB  
Article
Identification and Characterization of Fully Human FOLR1-Targeting CAR T Cells for the Treatment of Ovarian Cancer
by Maria Bethke, Pierre Abramowski, Miriam Droste, André Felsberger, Lisa Kochsiek, Bettina Kotter, Luisa Plettig, Kateryna Antonova, Salpy Baghdo, Nico Burzan, Florian Tomszak, Manuel Martinez-Osuna, Dominik Eckardt and Christoph Herbel
Cells 2024, 13(22), 1880; https://doi.org/10.3390/cells13221880 - 14 Nov 2024
Viewed by 517
Abstract
CAR T cell therapy has been an effective treatment option for hematological malignancies. However, the therapeutic potential of CAR T cells can be reduced by several constraints, partly due to immunogenicity and toxicities. The lack of established workflows enabling thorough evaluation of new [...] Read more.
CAR T cell therapy has been an effective treatment option for hematological malignancies. However, the therapeutic potential of CAR T cells can be reduced by several constraints, partly due to immunogenicity and toxicities. The lack of established workflows enabling thorough evaluation of new candidates, limits comprehensive CAR assessment. To improve the selection of lead CAR candidates, we established a stringent, multistep workflow based on specificity assessments, employing multiple assays and technologies. Moreover, we characterized a human FOLR1-directed CAR binding domain. Selection of binding domains was based on extensive specificity assessment by flow cytometry and imaging, to determine on-/off-target and off-tumor reactivity. CAR T cell functionality and specificity were assessed by high-throughput screening and advanced in vitro assays. Our validation strategy highlights that assays comprehensively characterizing CAR functionality and binding specificity complement each other. Thereby, critical specificity considerations can be addressed early in the development process to overcome current limitations for future CAR T cell therapies. Full article
Show Figures

Figure 1

19 pages, 1526 KiB  
Review
The Role of Adenosine in Overcoming Resistance in Sarcomas
by Marlid Cruz-Ramos, Sara Aileen Cabrera-Nieto, Mario Murguia-Perez and Fernanda Sarahí Fajardo-Espinoza
Int. J. Mol. Sci. 2024, 25(22), 12209; https://doi.org/10.3390/ijms252212209 - 14 Nov 2024
Viewed by 253
Abstract
Resistance to systemic therapies in sarcomas poses a significant challenge to improving clinical outcomes. Recent research has concentrated on the tumor microenvironment’s role in sarcoma progression and treatment resistance. This microenvironment comprises a variety of cell types and signaling molecules that influence tumor [...] Read more.
Resistance to systemic therapies in sarcomas poses a significant challenge to improving clinical outcomes. Recent research has concentrated on the tumor microenvironment’s role in sarcoma progression and treatment resistance. This microenvironment comprises a variety of cell types and signaling molecules that influence tumor behavior, including proliferation, metastasis, and resistance to therapy. Adenosine, abundant in the tumor microenvironment, has been implicated in promoting immunosuppression and chemoresistance. Targeting adenosine receptors and associated pathways offers a novel approach to enhancing immune responses against tumors, potentially improving immunotherapy outcomes in cancers, including sarcomas. Manipulating adenosine signaling also shows promise in overcoming chemotherapy resistance in these tumors. Clinical trials investigating adenosine receptor antagonists in sarcomas have fueled interest in this pathway for sarcoma treatment. Ultimately, a comprehensive understanding of the tumor and vascular microenvironments, as well as the adenosine pathway, may open new avenues for improving treatment outcomes and overcoming resistance in sarcoma. Further studies and clinical trials are crucial to validate these findings and optimize therapeutic strategies, particularly for osteosarcoma. This study provides a literature review exploring the potential role of the adenosine pathway in sarcomas. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

23 pages, 1586 KiB  
Review
Oncolytic Virotherapies and Adjuvant Gut Microbiome Therapeutics to Enhance Efficacy Against Malignant Gliomas
by Natalie M. Meléndez-Vázquez, Candelaria Gomez-Manzano and Filipa Godoy-Vitorino
Viruses 2024, 16(11), 1775; https://doi.org/10.3390/v16111775 - 14 Nov 2024
Viewed by 518
Abstract
Glioblastoma (GBM) is the most prevalent malignant brain tumor. Current standard-of-care treatments offer limited benefits for patient survival. Virotherapy is emerging as a novel strategy to use oncolytic viruses (OVs) for the treatment of GBM. These engineered and non-engineered viruses infect and lyse [...] Read more.
Glioblastoma (GBM) is the most prevalent malignant brain tumor. Current standard-of-care treatments offer limited benefits for patient survival. Virotherapy is emerging as a novel strategy to use oncolytic viruses (OVs) for the treatment of GBM. These engineered and non-engineered viruses infect and lyse cancer cells, causing tumor destruction without harming healthy cells. Recent advances in genetic modifications to OVs have helped improve their targeting capabilities and introduce therapeutic genes, broadening the therapeutic window and minimizing potential side effects. The efficacy of oncolytic virotherapy can be enhanced by combining it with other treatments such as immunotherapy, chemotherapy, or radiation. Recent studies suggest that manipulating the gut microbiome to enhance immune responses helps improve the therapeutic efficacy of the OVs. This narrative review intends to explore OVs and their role against solid tumors, especially GBM while emphasizing the latest technologies used to enhance and improve its therapeutic and clinical responses. Full article
(This article belongs to the Special Issue Progress and Prospects in Oncolytic Virotherapy)
Show Figures

Figure 1

15 pages, 2409 KiB  
Systematic Review
Risk Factors of Immune-Mediated Hepatotoxicity Induced by Immune Checkpoint Inhibitors in Cancer Patients: A Systematic Review and Meta-Analysis
by Ying Jiang, Ranyi Li, Xiaoyu Li and Ningping Zhang
Curr. Oncol. 2024, 31(11), 7129-7143; https://doi.org/10.3390/curroncol31110525 - 13 Nov 2024
Viewed by 422
Abstract
Immune checkpoint inhibitors (ICIs) significantly improve survival, while immune-mediated hepatotoxicity (IMH) has been reported. To evaluate the incidence and potential risk factors of IMH among cancer patients treated by ICIs, PubMed/Medline, Web of Science, Cochrane, and Embase were searched before 30 March 2024 [...] Read more.
Immune checkpoint inhibitors (ICIs) significantly improve survival, while immune-mediated hepatotoxicity (IMH) has been reported. To evaluate the incidence and potential risk factors of IMH among cancer patients treated by ICIs, PubMed/Medline, Web of Science, Cochrane, and Embase were searched before 30 March 2024 for systematic review and meta-analysis. Odds ratios (ORs) with 95% confidence intervals (CI) were calculated. Quality assessment was completed using the Newcastle–Ottawa scale. Of 1217 articles identified, 24 consisting of 9076 patients were included, with one study being prospective and the rest retrospective. The overall incidence of any grade IMH and grade ≥ 3 secondary to ICIs was 14% and 7%, respectively. The cholestatic pattern was more prevalent than the hepatocellular and mixed patterns. The meta-analysis revealed that ICI treatment was related to reduced risk of IMH in older patients (SMD: −0.18; 95% CI: −0.33 to −0.04), individuals with higher body mass index (WMD: −2.15; 95% CI: −3.92 to −0.38), males (OR: 0.44; 95% CI: 0.27 to 0.72), and patients with lung cancer (OR: 0.58, 95%CI 0.41 to 0.83). On the other hand, patients with liver metastasis (OR: 1.80; 95% CI: 1.47 to 2.20), history of ICI treatment (OR: 3.09; 95% CI: 1.21 to 7.89), diabetes (OR: 2.19; 95% CI: 1.36 to 3.51), chronic HBV (OR: 3.06; 95% CI: 1.11 to 8.46), and concomitant use of ICIs (OR: 8.73; 95% CI: 2.41 to 31.59) increased the risk of developing IMH. This study will provide clinicians with information on potentially high-risk groups for IMH, who need to be cautiously monitored for liver function when receiving immunotherapy. Full article
Show Figures

Figure 1

Back to TopTop