[go: up one dir, main page]

 
 
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,720)

Search Parameters:
Keywords = HCC

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 1043 KiB  
Review
Research Progress on Dendritic Cells in Hepatocellular Carcinoma Immune Microenvironments
by Wenya Li, Guojie Chen, Hailin Peng, Qingfang Zhang, Dengyun Nie, Ting Guo, Yinxing Zhu, Yuhan Zhang and Mei Lin
Biomolecules 2024, 14(9), 1161; https://doi.org/10.3390/biom14091161 - 16 Sep 2024
Abstract
Dendritic cells (DCs) are antigen-presenting cells that play a crucial role in initiating immune responses by cross-presenting relevant antigens to initial T cells. The activation of DCs is a crucial step in inducing anti-tumor immunity. Upon recognition and uptake of tumor antigens, activated [...] Read more.
Dendritic cells (DCs) are antigen-presenting cells that play a crucial role in initiating immune responses by cross-presenting relevant antigens to initial T cells. The activation of DCs is a crucial step in inducing anti-tumor immunity. Upon recognition and uptake of tumor antigens, activated DCs present these antigens to naive T cells, thereby stimulating T cell-mediated immune responses and enhancing their ability to attack tumors. It is particularly noted that DCs are able to cross-present foreign antigens to major histocompatibility complex class I (MHC-I) molecules, prompting CD8+ T cells to proliferate and differentiate into cytotoxic T cells. In the malignant progression of hepatocellular carcinoma (HCC), the inactivation of DCs plays an important role, and the activation of DCs is particularly important in anti-HCC immunotherapy. In this review, we summarize the mechanisms of DC activation in HCC, the involved regulatory factors and strategies to activate DCs in HCC immunotherapy. It provides a basis for the study of HCC immunotherapy through DC activation. Full article
25 pages, 8440 KiB  
Review
Targeted Drug Delivery Strategies for the Treatment of Hepatocellular Carcinoma
by Yonghui Liu, Yanan Wu, Zijian Li, Dong Wan and Jie Pan
Molecules 2024, 29(18), 4405; https://doi.org/10.3390/molecules29184405 - 16 Sep 2024
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent malignant tumors, exhibiting a high incidence rate that presents a substantial threat to human health. The use of sorafenib and lenvatinib, commonly employed as single-agent targeted inhibitors, complicates the treatment process due to the absence [...] Read more.
Hepatocellular carcinoma (HCC) ranks among the most prevalent malignant tumors, exhibiting a high incidence rate that presents a substantial threat to human health. The use of sorafenib and lenvatinib, commonly employed as single-agent targeted inhibitors, complicates the treatment process due to the absence of definitive targeting. Nevertheless, the advent of nanotechnology has injected new optimism into the domain of liver cancer therapy. Nanocarriers equipped with active targeting or passive targeting mechanisms have demonstrated the capability to deliver drugs to tumor cells with high efficiency. This approach not only facilitates precise delivery to the affected site but also enables targeted drug release, thereby enhancing therapeutic efficacy. As medical technology progresses, there is an increasing call for innovative treatment modalities, including novel chemotherapeutic agents, gene therapy, phototherapy, immunotherapy, and combinatorial treatments for HCC. These emerging therapies are anticipated to yield improved clinical outcomes for patients, while minimizing systemic toxicity and adverse effects. Consequently, the application of nanotechnology is poised to significantly improve HCC treatment. This review focused on targeted strategies for HCC and the application of nanotechnology in this area. Full article
Show Figures

Figure 1

Figure 1
<p>Treatment of HCC by targeting AFP [<a href="#B42-molecules-29-04405" class="html-bibr">42</a>]. Reprinted with permission from [<a href="#B42-molecules-29-04405" class="html-bibr">42</a>]. Copyright 2022, The American Association for the Advancement of Science.</p>
Full article ">Figure 2
<p>The mechanism of action of UR@M NPs for HCC immunotherapy. (<b>A</b>) Engineering of bionic UR@M NPs incorporating UA and CRISPR technologies. (<b>B</b>) UA combined with PD-L1 gene therapy to enhance immunotherapy with nanomedicines [<a href="#B67-molecules-29-04405" class="html-bibr">67</a>]. Reprinted with permission from [<a href="#B67-molecules-29-04405" class="html-bibr">67</a>]. Copyright 2024, Elsevier.</p>
Full article ">Figure 3
<p>Construction strategy of TCLMs and schematic diagram of target-generating chemo-photodynamic therapy [<a href="#B76-molecules-29-04405" class="html-bibr">76</a>]. Reprinted with permission from [<a href="#B76-molecules-29-04405" class="html-bibr">76</a>]. Copyright 2021, Elsevier.</p>
Full article ">Figure 4
<p>Schematic representation of MnO<sub>2</sub>/BPD synthesis and intervention in PDT-induced thrombosis. (<b>A</b>) After synthesized in vitro, the MnO<sub>2</sub>/BPD nanoparticle was injected to undergo the reduction and self-assemble in vivo. (<b>B</b>) Schematic representation of IPDT under the guidance of ultrasound [<a href="#B113-molecules-29-04405" class="html-bibr">113</a>]. Reprinted with permission from [<a href="#B113-molecules-29-04405" class="html-bibr">113</a>]. Copyright 2020, American Chemical Society.</p>
Full article ">Figure 5
<p>NIR-II photoacoustically guided controlled oxygenation bio-nanoparticles for highly specific HCC photodynamic therapy [<a href="#B158-molecules-29-04405" class="html-bibr">158</a>]. Reprinted with permission from [<a href="#B158-molecules-29-04405" class="html-bibr">158</a>]. Copyright 2024, Wiley.</p>
Full article ">Figure 6
<p>Supramolecular nanocarrier T-SPM<sub>DCTBT/NO</sub> for ONOO<sup>-</sup>-enhanced mesophilic PTT in HCC. (<b>A</b>) The basic chemical structures of the components of nanocarrier. (<b>B</b>) Schematic illustration of the supramolecular nanocarrier T-SPM<sub>DCTBT/NO</sub> for ONOO<sup>-</sup>-potentiated mild-temperature PTT of HCC [<a href="#B165-molecules-29-04405" class="html-bibr">165</a>]. Reprinted with permission from [<a href="#B165-molecules-29-04405" class="html-bibr">165</a>]. Copyright 2023, Wiley.</p>
Full article ">Figure 7
<p>Mechanisms of immune–gene therapy with siRNA-containing TT-LDCP NPs against the immune checkpoint PD-L1 and pDNA encoding the immunostimulatory cytokine IL-2 [<a href="#B172-molecules-29-04405" class="html-bibr">172</a>]. Reprinted with permission from [<a href="#B172-molecules-29-04405" class="html-bibr">172</a>]. Copyright 2020, The American Association for the Advancement of Science.</p>
Full article ">Figure 8
<p>Schematic diagram of BEA-C=N-DOX-M for synergistic chemoimmunotherapy of hepatocellular carcinoma. (<b>A</b>) The synthesis process of BEA-C=N-DOX-M. (<b>B</b>) The action process of this nano-micelles in vivo and the strategy for achieving chemo-immunotherapy in HCC [<a href="#B190-molecules-29-04405" class="html-bibr">190</a>]. Reprinted with permission from [<a href="#B190-molecules-29-04405" class="html-bibr">190</a>]. Copyright 2023, Wiley.</p>
Full article ">Scheme 1
<p>Targeted strategies for treating HCC. This diagram was drawn in Figdraw.</p>
Full article ">
19 pages, 1153 KiB  
Review
Biomarkers for Immunotherapy Efficacy in Advanced Hepatocellular Carcinoma: A Comprehensive Review
by Erfan Taherifard, Krystal Tran, Ali Saeed, Jehad Amer Yasin and Anwaar Saeed
Diagnostics 2024, 14(18), 2054; https://doi.org/10.3390/diagnostics14182054 - 16 Sep 2024
Viewed by 1
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver malignancy and the sixth most common cancer globally, remains fatal for many patients with inappropriate responses to treatment. Recent advancements in immunotherapy have transformed the treatment landscape for advanced HCC. However, variability in patient responses [...] Read more.
Hepatocellular carcinoma (HCC), the most common primary liver malignancy and the sixth most common cancer globally, remains fatal for many patients with inappropriate responses to treatment. Recent advancements in immunotherapy have transformed the treatment landscape for advanced HCC. However, variability in patient responses to immunotherapy highlights the need for biomarkers that can predict treatment outcomes. This manuscript comprehensively reviews the evolving role of biomarkers in immunotherapy efficacy, spanning from blood-derived indicators—alpha-fetoprotein, inflammatory markers, cytokines, circulating tumor cells, and their DNA—to tissue-derived indicators—programmed cell death ligand 1 expression, tumor mutational burden, microsatellite instability, and tumor-infiltrating lymphocytes. The current body of evidence suggests that these biomarkers hold promise for improving patient selection and predicting immunotherapy outcomes. Each biomarker offers unique insights into disease biology and the immune landscape of HCC, potentially enhancing the precision of treatment strategies. However, challenges such as methodological variability, high costs, inconsistent findings, and the need for large-scale validation in well-powered two-arm trial studies persist, making them currently unsuitable for integration into standard care. Addressing these challenges through standardized techniques and implementation of further studies will be critical for the future incorporation of these biomarkers into clinical practice for advanced HCC. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Overview of blood-derived and tissue-derived biomarkers in hepatocellular carcinoma immunotherapy. AFP: alpha-fetoprotein; NLR: neutrophil-to-lymphocyte ratio; PLR: platelet-to-lymphocyte ratio; ctDNA: circulating tumor DNA; CTC: circulating tumor cell; MSI: microsatellite instability; TIL: tumor-infiltrating lymphocyte.</p>
Full article ">Figure 2
<p>Circulating tumor DNA (CtDNA) and circulating tumor cells (CTCs) as liquid biopsy blood-derived biomarkers for immunotherapy response in hepatocellular carcinoma.</p>
Full article ">
16 pages, 1777 KiB  
Article
Metabolomics Biomarker Discovery to Optimize Hepatocellular Carcinoma Diagnosis: Methodology Integrating AutoML and Explainable Artificial Intelligence
by Fatma Hilal Yagin, Radwa El Shawi, Abdulmohsen Algarni, Cemil Colak, Fahaid Al-Hashem and Luca Paolo Ardigò
Diagnostics 2024, 14(18), 2049; https://doi.org/10.3390/diagnostics14182049 - 15 Sep 2024
Viewed by 235
Abstract
Background: This study aims to assess the efficacy of combining automated machine learning (AutoML) and explainable artificial intelligence (XAI) in identifying metabolomic biomarkers that can differentiate between hepatocellular carcinoma (HCC) and liver cirrhosis in patients with hepatitis C virus (HCV) infection. Methods: We [...] Read more.
Background: This study aims to assess the efficacy of combining automated machine learning (AutoML) and explainable artificial intelligence (XAI) in identifying metabolomic biomarkers that can differentiate between hepatocellular carcinoma (HCC) and liver cirrhosis in patients with hepatitis C virus (HCV) infection. Methods: We investigated publicly accessible data encompassing HCC patients and cirrhotic controls. The TPOT tool, which is an AutoML tool, was used to optimize the preparation of features and data, as well as to select the most suitable machine learning model. The TreeSHAP approach, which is a type of XAI, was used to interpret the model by assessing each metabolite’s individual contribution to the categorization process. Results: TPOT had superior performance in distinguishing between HCC and cirrhosis compared to other AutoML approaches AutoSKlearn and H2O AutoML, in addition to traditional machine learning models such as random forest, support vector machine, and k-nearest neighbor. The TPOT technique attained an AUC value of 0.81, showcasing superior accuracy, sensitivity, and specificity in comparison to the other models. Key metabolites, including L-valine, glycine, and DL-isoleucine, were identified as essential by TPOT and subsequently verified by TreeSHAP analysis. TreeSHAP provided a comprehensive explanation of the contribution of these metabolites to the model’s predictions, thereby increasing the interpretability and dependability of the results. This thorough assessment highlights the strength and reliability of the AutoML framework in the development of clinical biomarkers. Conclusions: This study shows that AutoML and XAI can be used together to create metabolomic biomarkers that are specific to HCC. The exceptional performance of TPOT in comparison to traditional models highlights its capacity to identify biomarkers. Furthermore, TreeSHAP boosted model transparency by highlighting the relevance of certain metabolites. This comprehensive method has the potential to enhance the identification of biomarkers and generate precise, easily understandable, AI-driven solutions for diagnosing HCC. Full article
Show Figures

Figure 1

Figure 1
<p>A diagram of the proposed method in the current research.</p>
Full article ">Figure 2
<p>Nemenyi Test (α = 0.05) comparing the AUC of testing data for AutoML techniques and traditional machine learning techniques.</p>
Full article ">Figure 3
<p>Feature importance ranking based on SHAP values.</p>
Full article ">Figure 4
<p>SHAP waterfall plot for a representative true positive sample.</p>
Full article ">Figure 5
<p>SHAP waterfall plot for a representative true negative sample.</p>
Full article ">Figure 6
<p>Partial dependence plot of L-valine 1 showing its SHAP value and interaction with 2,3-butanediol 2.</p>
Full article ">
11 pages, 642 KiB  
Article
Late Hepatocellular Carcinoma Occurrence in Patients Achieving Sustained Virological Response After Direct-Acting Antiviral Therapy: A Matter of Follow-Up or Something Else?
by Alessandro Perrella, Alfredo Caturano, Ilario de Sio, Pasquale Bellopede, Adelaide Maddaloni, Luigi Maria Vitale, Barbara Rinaldi, Andrea Mormone, Antonio Izzi, Costanza Sbreglia, Francesca Futura Bernardi, Ugo Trama, Massimiliano Berretta, Raffaele Galiero, Erica Vetrano, Ferdinando Carlo Sasso, Gianluigi Franci, Raffaele Marfella and Luca Rinaldi
J. Clin. Med. 2024, 13(18), 5474; https://doi.org/10.3390/jcm13185474 - 14 Sep 2024
Viewed by 404
Abstract
Background: Despite achieving a sustained virological response (SVR) with direct-acting antivirals (DAAs), an unexpected increase in the occurrence rate of hepatocellular carcinoma (HCC) has been observed among HCV-treated patients. This study aims to assess the long-term follow-up of HCV patients treated with [...] Read more.
Background: Despite achieving a sustained virological response (SVR) with direct-acting antivirals (DAAs), an unexpected increase in the occurrence rate of hepatocellular carcinoma (HCC) has been observed among HCV-treated patients. This study aims to assess the long-term follow-up of HCV patients treated with DAAs who achieved an SVR to investigate the potential for late-onset HCC. Methods: In this prospective multicenter study, we enrolled consecutive HCV patients treated with DAAs following Italian ministerial guidelines between 2015 and 2018. Exclusion criteria included active HCC on imaging, prior HCC treatment, HBV or HIV co-infection, or liver transplant recipients. Monthly follow-ups occurred during treatment, with subsequent assessments every 3 months for at least 48 months. Abdominal ultrasound (US) was performed within two weeks before starting antiviral therapy, supplemented by contrast-enhanced ultrasonography (CEUS), dynamic computed tomography (CT), or magnetic resonance imaging (MRI) to evaluate incidental liver lesions. Results: Of the 306 patients completing the 48-months follow-up post-treatment (median age 67 years, 55% male), all achieved an SVR. A sofosbuvir-based regimen was administered to 72.5% of patients, while 20% received ribavirin. During follow-up, late-onset HCC developed in 20 patients (cumulative incidence rate of 6.55%). The pattern of HCC occurrence varied (median diameter 24 mm). Multivariate and univariate analyses identified liver stiffness, diabetes, body mass index, and platelet levels before antiviral therapy as associated factors for late HCC occurrence. Conclusions: Our findings suggest that late HCC occurrence may persist despite achieving SVR. Therefore, comprehensive long-term follow-up, including clinical, laboratory, and expert ultrasonography evaluations, is crucial for all HCV patients treated with DAAs. Full article
Show Figures

Figure 1

Figure 1
<p>ROC curve describing the discriminant power of the liver stiffness value (kPa) based on the risk of developing late HCC in cirrhotic patients [<span class="html-italic">n</span> = 306, AUROC score = 0.646, 95% C.I.: 0.519–0.774]. The <span class="html-italic">p</span> value for the significance of liver stiffness based on the risk of HCC was 0.029 (Kruskal–Wallis test).</p>
Full article ">
21 pages, 1365 KiB  
Review
Recent Advances and Mechanisms of Phage-Based Therapies in Cancer Treatment
by Vivian Y. Ooi and Ting-Yu Yeh
Int. J. Mol. Sci. 2024, 25(18), 9938; https://doi.org/10.3390/ijms25189938 (registering DOI) - 14 Sep 2024
Viewed by 526
Abstract
The increasing interest in bacteriophage technology has prompted its novel applications to treat different medical conditions, most interestingly cancer. Due to their high specificity, manipulability, nontoxicity, and nanosize nature, phages are promising carriers in targeted therapy and cancer immunotherapy. This approach is particularly [...] Read more.
The increasing interest in bacteriophage technology has prompted its novel applications to treat different medical conditions, most interestingly cancer. Due to their high specificity, manipulability, nontoxicity, and nanosize nature, phages are promising carriers in targeted therapy and cancer immunotherapy. This approach is particularly timely, as current challenges in cancer research include damage to healthy cells, inefficiency in targeting, obstruction by biological barriers, and drug resistance. Some cancers are being kept at the forefront of phage research, such as colorectal cancer and HCC, while others like lymphoma, cervical cancer, and myeloma have not been retouched in a decade. Common mechanisms are immunogenic antigen display on phage coats and the use of phage as transporters to carry drugs, genes, and other molecules. To date, popular phage treatments being tested are gene therapy and phage-based vaccines using M13 and λ phage, with some vaccines having advanced to human clinical trials. The results from most of these studies have been promising, but limitations in phage-based therapies such as reticuloendothelial system clearance or diffusion inefficiency must be addressed. Before phage-based therapies for cancer can be successfully used in oncology practice, more in-depth research and support from local governments are required. Full article
(This article belongs to the Special Issue Bacteriophages Biology and Bacteriophage-Derived Technologies)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Mechanisms of phage-based cancer therapies (using filamentous phage as an example). (<b>A</b>) Cancer cell antigens can be displayed on phage surface using peptide display technique. These antigens can trigger an immune response, encourage production of anti-cancer antibodies, or activate cytotoxic T cells against cancer cells. (<b>B</b>) Phages can be used as transporters to deliver photosensitizers, cytokines, or transgenes to cancer cells. APC, antigen presenting cell; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand; pIII, M13 pIII coat protein; pVIII, M13 pVIII coat protein.</p>
Full article ">
11 pages, 3964 KiB  
Article
Adverse Events in Targeted Therapy for Unresectable Hepatocellular Carcinoma Predict Clinical Outcomes
by Kenji Imai, Koji Takai, Masashi Aiba, Shinji Unome, Takao Miwa, Tatsunori Hanai, Atsushi Suetsugu and Masahito Shimizu
Cancers 2024, 16(18), 3150; https://doi.org/10.3390/cancers16183150 - 14 Sep 2024
Viewed by 247
Abstract
To assess the impact of adverse event (AE) severity, caused by targeted therapy, on overall survival (OS) and progression-free survival (PFS) in patients with unresectable hepatocellular carcinoma (HCC), a total of 183 patients with HCC treated with atezolizumab plus bevacizumab (40), lenvatinib (57), [...] Read more.
To assess the impact of adverse event (AE) severity, caused by targeted therapy, on overall survival (OS) and progression-free survival (PFS) in patients with unresectable hepatocellular carcinoma (HCC), a total of 183 patients with HCC treated with atezolizumab plus bevacizumab (40), lenvatinib (57), sorafenib (79), cabozantinib (3), ramucirumab (3), and regorafenib (1) were included in this study. Age-, AFP-, and ALBI score-adjusted hazard ratios (HRs) of AE grades 1 to 3 versus grade 0 for OS and PFS were calculated using Cox proportional hazards models. The linear trend of the HRs was assessed by calculating the p values for this trend. The most common AEs were appetite loss (AE grade 0/1/2/3 = 97/23/55/12), general fatigue (102/31/44/6), hypertension (120/6/40/17), hand-foot syndrome (HFS) (135/21/24/3), proteinuria (140/13/16/14), and hypothyroidism (148/12/23/0). The adjusted HRs for OS of these AEs were 0.532–1.450–2.361 (p for trend 0.037), 1.057–1.691–3.364 (p for trend 0.004), 1.176–0.686–0.281 (p for trend 0.002), 0.639–0.759–1.820 (p for trend 0.462), 1.030–0.959–0.147 (p for trend 0.011), and 0.697–0.609 (p for trend 0.119), respectively. Those for PFS of the corresponding AEs were 0.592–1.073–2.811 (p for trend 0.255), 1.161–1.282–4.324 (p for trend 0.03), 0.965–0.781–0.655 (p for trend 0.095), 0.737–0.623–2.147 (p for trend 0.153), 1.061–0.832–0.800 (p for trend 0.391), and 1.412–0.560 (p for trend 0.081), respectively. Appetite loss and general fatigue negatively affected clinical outcomes, whereas hypertension, HFS, proteinuria, and hypothyroidism had positive effects. Full article
Show Figures

Figure 1

Figure 1
<p>Overall survival (OS) curves for each grade of targeted therapy-induced adverse event. The differences between the survival curves (<b>a</b>–<b>f</b>) were assessed using the log-rank test, with multiple comparisons adjusted via Bonferroni correction.</p>
Full article ">Figure 2
<p>Hazard ratios for overall survival comparing adverse event grades 1 to 3 with 0. * Adjusted for age, alpha-fetoprotein, and ALBI score.</p>
Full article ">Figure 3
<p>Progression-free survival (PFS) curves for each grade of targeted therapy-induced adverse event. The differences between the survival curves (<b>a</b>–<b>f</b>) were assessed using the log-rank test, with multiple comparisons adjusted via Bonferroni correction.</p>
Full article ">Figure 4
<p>Hazard ratios for progression-free survival comparing adverse event grades 1 to 3 with 0. * Adjusted for age, alpha-fetoprotein, and ALBI score.</p>
Full article ">
24 pages, 1170 KiB  
Review
Understanding Macrophage Complexity in Metabolic Dysfunction-Associated Steatotic Liver Disease: Transitioning from the M1/M2 Paradigm to Spatial Dynamics
by Forkan Ahamed, Natalie Eppler, Elizabeth Jones and Yuxia Zhang
Livers 2024, 4(3), 455-478; https://doi.org/10.3390/livers4030033 - 13 Sep 2024
Viewed by 249
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses metabolic dysfunction-associated fatty liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), with MASH posing a risk of progression to cirrhosis and hepatocellular carcinoma (HCC). The global prevalence of MASLD is estimated at approximately a quarter of the [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses metabolic dysfunction-associated fatty liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), with MASH posing a risk of progression to cirrhosis and hepatocellular carcinoma (HCC). The global prevalence of MASLD is estimated at approximately a quarter of the population, with significant healthcare costs and implications for liver transplantation. The pathogenesis of MASLD involves intrahepatic liver cells, extrahepatic components, and immunological aspects, particularly the involvement of macrophages. Hepatic macrophages are a crucial cellular component of the liver and play important roles in liver function, contributing significantly to tissue homeostasis and swift responses during pathophysiological conditions. Recent advancements in technology have revealed the remarkable heterogeneity and plasticity of hepatic macrophage populations and their activation states in MASLD, challenging traditional classification methods like the M1/M2 paradigm and highlighting the coexistence of harmful and beneficial macrophage phenotypes that are dynamically regulated during MASLD progression. This complexity underscores the importance of considering macrophage heterogeneity in therapeutic targeting strategies, including their distinct ontogeny and functional phenotypes. This review provides an overview of macrophage involvement in MASLD progression, combining traditional paradigms with recent insights from single-cell analysis and spatial dynamics. It also addresses unresolved questions and challenges in this area. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic overview of hepatic sinusoid and macrophages. The liver is divided into three zones: the areas around the hepatic arteries and portal veins are known as zone 1, those near the central vein are zone 3, and the cells in between are referred to as zone 2. Oxygen-rich blood from the hepatic artery combines with nutrient-rich blood from the portal vein and flows along the sinusoids toward the central vein (red arrow). Meanwhile, bile flows from zone 3 to zone 1, collected by the bile ducts (green arrow). Hepatic macrophages consist primarily of two distinct subtypes: liver resident Kupffer cells (KC), originating from yolk sac, and monocyte-derived macrophages (MDM), from the bone marrow (black arrow). KCs and MDMs can be differentiated by their distinct cell surface markers. Located near liver sinusoidal endothelial cells (LSECs) along the hepatic sinusoids, KCs and MDMs play an important role in influencing the activity of hepatic stellate cells (HSCs) and hepatocytes.</p>
Full article ">Figure 2
<p>Schematic overview of macrophage M1 and M2 polarization. Macrophages can polarize into two distinct phenotypes depending on the microenvironmental stimuli they encounter. M1 macrophages, induced by LPS and IFN-γ, activate pathways such as TLR/NF-κB, STAT1, NOTCH, mTOR/PI3K/Akt, and JNK/c-Myc, leading to the release of pro-inflammatory factors like IL-1, IL-12, IL-18, iNOS, and TNF-α. In contrast, M2 macrophages, induced by IL-4, IL-10, and IL-13, promote anti-inflammatory activity through pathways like STAT3/6, TGF-β/SMADs, and PPAR (α, β/δ, and γ), expressing IL-10 and arginase 1.</p>
Full article ">
10 pages, 1735 KiB  
Article
Low CD8+ Density Variation and R1 Surgical Margin as Independent Predictors of Early Post-Resection Recurrence in HCC Patients Meeting Milan Criteria
by Rokas Stulpinas, Ieva Jakiunaite, Agne Sidabraite, Allan Rasmusson, Dovile Zilenaite-Petrulaitiene, Kestutis Strupas, Arvydas Laurinavicius and Aiste Gulla
Curr. Oncol. 2024, 31(9), 5344-5353; https://doi.org/10.3390/curroncol31090394 - 10 Sep 2024
Viewed by 237
Abstract
Our study included 41 patients fulfilling the Milan criteria preoperatively and aimed to identify individuals at high risk of post-resection HCC relapse, which occurred in 18 out of 41 patients (43.9%), retrospectively. We analyzed whole slide images of CD8 immunohistochemistry with automated segmentation [...] Read more.
Our study included 41 patients fulfilling the Milan criteria preoperatively and aimed to identify individuals at high risk of post-resection HCC relapse, which occurred in 18 out of 41 patients (43.9%), retrospectively. We analyzed whole slide images of CD8 immunohistochemistry with automated segmentation of tissue classes and detection of CD8+ lymphocytes. The image analysis outputs were subsampled using a hexagonal grid-based method to assess spatial distribution of CD8+ lymphocytes with regards to the epithelial edges. The CD8+ lymphocyte density indicators, along with clinical, radiological, post-surgical and pathological variables, were tested to predict HCC relapse. Low standard deviation of CD8+ density along the tumor edge and R1 resection emerged as independent predictors of shorter recurrence-free survival (RFS). In particular, patients presenting with both adverse predictors exhibited 100% risk of relapse within 200 days. Our results highlight the potential utility of integrating CD8+ density variability and surgical margin to identify a high relapse-risk group among Milan criteria-fulfilling HCC patients. Validation in cohorts with core biopsy could provide CD8+ distribution data preoperatively and guide preoperative decisions, potentially prioritizing liver transplantation for patients at risk of incomplete resection (R1) and thereby improving overall treatment outcomes significantly. Full article
(This article belongs to the Special Issue Novel Biomarkers and Liver Cancer)
Show Figures

Figure 1

Figure 1
<p>Study workflow. Pre-analytical step: HCC patients meeting the Milan criteria underwent liver resection between 2007–2020. Samples were analyzed and scanned slides were annotated by a pathologist. Digital image analysis: tissue samples were segmented into epithelial and stromal classes, and CD8+ lymphocytes were detected using HALO<sup>®</sup>AI (Indica Labs, USA). Hexagonal grid subsampling was applied for epithelial edge detection. Statistical analysis: aggregated CD8+ data per hexagon were combined with clinical data for prognostic modeling. Independent predictors were analyzed, and a relapse risk score was developed to prioritize liver transplant candidates.</p>
Full article ">Figure 2
<p>The R1 resection (<b>a</b>) and low standard deviation of CD8+ density along the tumor edge (<b>b</b>) as univariate predictors for shorter RFS.</p>
Full article ">Figure 3
<p>HCC Recurrence Risk Score: (<b>a</b>) 0 vs. 1–2 (<span class="html-italic">p</span> = 0.00026) and (<b>b</b>) 0–1 vs. 2 (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
11 pages, 1130 KiB  
Article
Prediction of Clinical Trajectory in HCV-Related ACLD after SVR: Role of Liver Stiffness in a 5-Years Prospective Study
by Filomena Morisco, Alessandro Federico, Massimo Marignani, Flavia L. Lombardo, Valentina Cossiga, Luisa Ranieri, Mario Romeo, Marina Cipullo, Paola Begini, Alessandra Zannella and Tommaso Stroffolini
Viruses 2024, 16(9), 1439; https://doi.org/10.3390/v16091439 - 10 Sep 2024
Viewed by 239
Abstract
The prediction of liver-related events (LRE) after sustained virological response (SVR) in HCV-advanced chronic liver disease (ACLD) patients is crucial. We aimed to evaluate incidence and risk factors of LRE in HCV-cirrhotic patients after SVR and to assess dynamic changes of liver stiffness [...] Read more.
The prediction of liver-related events (LRE) after sustained virological response (SVR) in HCV-advanced chronic liver disease (ACLD) patients is crucial. We aimed to evaluate incidence and risk factors of LRE in HCV-cirrhotic patients after SVR and to assess dynamic changes of liver stiffness in participants without LRE at the end of follow-up. We enrolled 575 consecutive patients with HCV-ACLD treated with DAAs and followed up for 5 years after SVR12. Overall, 98 (17%) patients developed any type of event, and HCC was the most frequent LRE. The incidence rate was 1.6 per 100 person-years (p/y) for both HCC and hepatic decompensation. Baseline LSM ≥ 20 kPa was the only independent predictor of hepatic decompensation, while LSM ≥ 20 kPa and male sex were independent predictors of HCC development. Among the 341 participants without LRE and with paired LSM, any LSM reduction was observed in 314 (92.1%), and half of them showed a decrease of LSM ≥ 20%. Among patients without LRE, 27.3% of participants without ≥20% LSM decrease at 2 years achieved the 5-year goal; in contrast, 31.6% of participants with ≥20% LSM decrease at 2 years lost it at 5 years. These findings provide evidence that baseline LSM is a tool to stratify patients at risk of developing LRE; the dynamic changes of LSM value suggest the need for monitoring this parameter over time. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

Figure 1
<p>Cumulative incidence of liver decompensation (<b>A</b>) and HCC (<b>B</b>) in cirrhotic patients according to the value of stiffness at baseline (Kaplan–Meier estimates).</p>
Full article ">Figure 2
<p>Dynamics of liver stiffness in patients without LRE at T24 and T60.</p>
Full article ">Figure 3
<p>Changes over time in fibrosis degree from 2 to 5 years of follow-up in SVR patients without LRE.</p>
Full article ">
11 pages, 980 KiB  
Article
The Real-World Efficacy and Safety of Direct-Acting Antivirals for Chronic Hepatitis C in Patients Active Malignancies
by Maria Dąbrowska, Jerzy Jaroszewicz, Marek Sitko, Justyna Janocha-Litwin, Dorota Zarębska-Michaluk, Ewa Janczewska, Beata Lorenc, Magdalena Tudrujek-Zdunek, Anna Parfieniuk-Kowerda, Jakub Klapaczyński, Hanna Berak, Łukasz Socha, Beata Dobracka, Dorota Dybowska, Włodzimierz Mazur, Łukasz Ważny and Robert Flisiak
Cancers 2024, 16(17), 3114; https://doi.org/10.3390/cancers16173114 - 9 Sep 2024
Viewed by 331
Abstract
Background: Over the past years, the introduction of direct-acting antivirals (DAAs) revolutionized chronic hepatitis C treatment. We aimed to characterize and assess treatment efficacy in three specific groups of patients treated with DAAs: those with active solid malignant tumors (SMTs), hematological diseases (HDs) [...] Read more.
Background: Over the past years, the introduction of direct-acting antivirals (DAAs) revolutionized chronic hepatitis C treatment. We aimed to characterize and assess treatment efficacy in three specific groups of patients treated with DAAs: those with active solid malignant tumors (SMTs), hematological diseases (HDs) and hepatocellular carcinomas (HCCs). Methods: A total of 203 patients with active oncological disease (SMT n = 61, HD = 67, HCC n = 74) during DAA treatment in 2015–2020 selected from the EpiTer-2 database were analyzed retrospectively and compared to 12,983 patients without any active malignancy. Results: Extrahepatic symptoms were more frequent in HD patients (17.2% vs. SMT = 10.3%, HCC = 8.2%, without = 7.8%, p = 0.004). HCC patients characterized with the highest ALT activity (81 IU/L vs. SMT = 59.5 IU/L, HD = 52 IU/L, without = 58 IU/L, p = 0.001) more often had F4 fibrosis as well (86.11% vs. SMT = 23.3%, HD = 28.8%, controls = 24.4%, p = 0.001). A significant majority of subjects in HCC, HD and SMT populations completed the full treatment plan (HCC = 91%; n = 67, HD = 97%; n = 65, SMT = 100%; n = 62). Concerning the treatment efficacy, the overall sustained virologic response, excluding non-virologic failures, was reported in 93.6% HD, 90.16% SMT and 80.6% in HCC patients. Conclusions: As presented in our study, DAA therapy has proven to be highly effective and safe in patients with active SMTs and HDs. However, therapy discontinuations resulting from liver disease progression remain to be the major concern in HCC patients. Full article
(This article belongs to the Special Issue Advances in the Prevention and Treatment of Liver Cancer)
Show Figures

Figure 1

Figure 1
<p>Flow chart showing the selection of patients included in this study.</p>
Full article ">Figure 2
<p>Efficacy of DAAs in patients with active oncological disease: SVR rates in ITT and PP analyses: excluding non-virologic failures. HCC, hepatocellular carcinoma; HD, hemato-oncologic disease; ITT, intention to treat; PP, per protocol; SMT, solid malignant tumor; SVR, sustained virologic response.</p>
Full article ">Figure 3
<p>Percentage of patients completing DAA therapy as recommended. DAA, direct-acting antiviral; HCC, hepatocellular carcinoma; HD, hemato-oncologic disease; SMT, solid malignant tumor.</p>
Full article ">
13 pages, 2231 KiB  
Article
Causal Effect of Immunocytes, Plasma Metabolites, and Hepatocellular Carcinoma: A Bidirectional Two-Sample Mendelian Randomization Study and Mediation Analysis in East Asian Populations
by Xilong Tang, Jianjin Xue, Jie Zhang and Jiajia Zhou
Genes 2024, 15(9), 1183; https://doi.org/10.3390/genes15091183 - 9 Sep 2024
Viewed by 348
Abstract
Background: Hepatocellular carcinoma (HCC) is a primary malignant liver tumor characterized by a low survival rate and high mortality. This study aimed to investigate the causal effect of immune cell phenotypes, plasma metabolites, and HCC in East Asian populations. Methods: The [...] Read more.
Background: Hepatocellular carcinoma (HCC) is a primary malignant liver tumor characterized by a low survival rate and high mortality. This study aimed to investigate the causal effect of immune cell phenotypes, plasma metabolites, and HCC in East Asian populations. Methods: The summary results for 731 immunocytes, 1400 plasma metabolites, and HCCs were acquired from publicly available genome-wide association studies (GWASs). This study utilized two-sample Mendelian randomization (MR) analysis to establish causal relationships, which was achieved by employing various statistical methods including inverse variance-weighted, simple mode, MR–Egger, weighted median, and weighted mode. Multiple sensitivity analyses were conducted to confirm the reliability of the MR data. Ultimately, mediation analysis was employed to ascertain the path that leads from immunocytes to plasma metabolites. Results: Among the 20 immune cells and HCC for East Asians, causal links were found, with one showing an inverse correlation. In addition, 36 metabolites were significantly associated with HCC for East Asians. Through analysis of established causative metabolites, we identified a strong correlation between the glycerophospholipid metabolic pathway and HCC for East Asians. By employing a two-step MR analysis, we identified 11 immunocytes that are causally linked to HCC for East Asians through the mediation of 14 plasma metabolites, with Linolenate [α or γ; (18:3n3 or 6)] levels showing the highest mediation proportion (19.3%). Conclusions: Our findings affirm the causal links among immunocytes, plasma metabolites, and HCC in eastern Asia populations by calculating the percentage of the impact that is influenced by plasma metabolites. This study offers innovative perspectives on the early detection, diagnosis, and therapy of HCC. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Flowchart of this research. HCC, hepatocellular carcinoma; MR, Mendelian randomization.</p>
Full article ">Figure 2
<p>Forest plots depicting the causal impacts of immunocytes on HCC. OR, odds ratio; CI, confidence interval.</p>
Full article ">Figure 3
<p>Forest plots depicting the causal impacts of plasma metabolites on HCC. ※ represents metabolites included in “glycerophospholipid metabolism”; OR, odds ratio; CI, confidence interval.</p>
Full article ">
16 pages, 8434 KiB  
Article
Thiostrepton as a Potential Therapeutic Agent for Hepatocellular Carcinoma
by Guifeng Su, Qianqing Yang, Heyang Zhou, Ying Huang, Shiyun Nie, Dan Wang, Guangchao Ma, Shaohua Zhang, Lingmei Kong, Chenggang Zou and Yan Li
Int. J. Mol. Sci. 2024, 25(17), 9717; https://doi.org/10.3390/ijms25179717 - 8 Sep 2024
Viewed by 330
Abstract
Due to limited drug efficacy and drug resistance, it is urgent to explore effective anti-liver cancer drugs. Repurposing drugs is an efficient strategy, with advantages including reduced costs, shortened development cycles, and assured safety. In this study, we adopted a synergistic approach combining [...] Read more.
Due to limited drug efficacy and drug resistance, it is urgent to explore effective anti-liver cancer drugs. Repurposing drugs is an efficient strategy, with advantages including reduced costs, shortened development cycles, and assured safety. In this study, we adopted a synergistic approach combining computational and experimental methods and identified the antibacterial drug thiostrepton (TST) as a candidate for an anti-liver cancer drug. Although the anti-tumor capabilities of TST have been reported, its role and underlying mechanisms in hepatocellular carcinoma (HCC) remain unclear. TST was found here to inhibit the proliferation of HCC cells effectively, arresting the cell cycle and inducing cell apoptosis, as well as suppressing the cell migration. Further, our findings revealed that TST induced mitochondrial impairment, which was demonstrated by destroyed mitochondrial structures, reduced mitochondria, and decreased mitochondrial membrane potential (MMP). TST caused the production of reactive oxygen species (ROS), and the mitochondrial impairment and proliferation inhibition of HCC cells were completely restored by the ROS scavenger N-acetyl-L-cysteine (NAC). Moreover, we discovered that TST induced mitophagy, and autophagy inhibition effectively promoted the anti-cancer effects of TST on HCC cells. In conclusion, our study suggests TST as a promising candidate for the treatment of liver cancers, and these findings provide theoretical support for the further development and potential application of TST in clinical liver cancer therapy. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>TST suppresses the proliferation of HCC. (<b>A</b>) Chemical structure of TST. (<b>B</b>) The cellular viability of HCC cells incubated with various concentrations of TST for 24 or 48 h was assessed using MTS solution. (<b>C</b>) IC<sub>50</sub> calculated from dose–response curves. (<b>D</b>) Colony formation in SK-Hep1 and HUH7 cells following 2 weeks of TST treatment. Quantification of cell colony numbers is shown on the right. *** <span class="html-italic">p</span> &lt; 0.001 versus the control group.</p>
Full article ">Figure 2
<p>TST suppresses the migration of HCC cells. (<b>A</b>) The migratory capacity of liver cancer cells treated with TST was assessed with cell scratch assay. Scale bar: 100 μm. (<b>B</b>) Statistical analysis of cell migration rate was displayed in histograms. * <span class="html-italic">p</span> &lt; 0.05, and *** <span class="html-italic">p</span> &lt; 0.001 versus the control group. (<b>C</b>) Western blot analysis revealed the expression levels of migration-related proteins, including N-cadherin, Snail, Slug, and Vimentin, in HCC cells subjected to TST treatment.</p>
Full article ">Figure 3
<p>TST induces apoptosis and arrests the cell cycle in HCC cells. (<b>A</b>) The cell phase distribution of HCC cells after 24 h of TST treatment, with the analysis results presented as histograms. ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 versus the control group. (<b>B</b>) Apoptosis of HCC cells treated with TST was assessed with flow cytometry. Statistical analysis of cell apoptosis rates was displayed in histograms. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 versus the control group. (<b>C</b>) Apoptosis-related proteins were detected by western blotting using proteins extracted from liver cancer cells treated with TST for 24 h.</p>
Full article ">Figure 4
<p>TST induces mitochondrial impairment in HCC cells. (<b>A</b>) Mitochondrial structure in TST-treated SK-Hep1 cells under electron microscope. The red arrows indicate mitochondria. (<b>B</b>) SK-Hep1 cells treated with TST for 24 h were stained with MitoTracker<sup>®</sup> Orange CMTMRos and subsequently observed under a fluorescence microscope. (<b>C</b>) The percentage of cells exhibiting low MitoTracker fluorescence was quantified using flow cytometry. (<b>D</b>) HCC cells treated with TST (4 μM) for the indicated time point were stained with JC-1 dye and subsequently observed under a fluorescence microscope. Red fluorescence signifies the accumulation of JC-1 in normal mitochondria, while green fluorescence indicates the presence of JC-1 monomers in the cellular matrix due to the reduction in MMP. (<b>E</b>) The MMP of SK-Hep1 and HUH7 cells treated with TST or the mitochondrial uncoupler CCCP for the indicated time point was assessed with flow cytometry. Changes in MMP are shown in histograms. Scale bar: 100 μm. ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 versus the control group. CCCP: carbonyl cyanide m-chlorophenyl hydrazone.</p>
Full article ">Figure 5
<p>TST-induced mitochondrial damage in HCC cells depends on ROS. (<b>A</b>,<b>B</b>) ROS in HCC cells treated with TST in the absence or presence of NAC for 24 h were assessed with flow cytometry. * <span class="html-italic">p</span> &lt; 0.05, and *** <span class="html-italic">p</span> &lt; 0.001 versus the control group. (<b>C</b>,<b>D</b>) HCC cells treated with TST in the absence or presence of NAC for 24 h were stained with MitoTracker<sup>®</sup> Orange CMTMRos and subsequently observed under a fluorescence microscope. (<b>E</b>,<b>F</b>) Quantification of the fluorescence intensity in the population of cells exhibiting low MitoTracker fluorescence by flow cytometry. Scale bar: 100 μm. NAC: N-acetyl-L-cysteine.</p>
Full article ">Figure 6
<p>ROS scavenging with NAC eliminates the inhibitory activity of TST against HCC cells. (<b>A</b>) Graphic representation of cell morphology after treatment with TST in the absence or presence of NAC (5 mM) for 24 h. (<b>B</b>) The cellular viability of HCC cells treated with TST in the absence or presence of NAC (5 mM) for 24 h was assessed using MTS solution. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 versus the control group. (<b>C</b>) Apoptosis of HCC cells treated with TST in the absence or presence of NAC (5 mM) for 24 h was assessed with flow cytometry. (<b>D</b>) Statistical analysis of cell apoptosis rate. Scale bar: 100 μm.</p>
Full article ">Figure 7
<p>Mitophagy blockade enhances the inhibitory activity of TST against HCC cells. (<b>A</b>) HCC cells treated with TST or CCCP for 12 h were stained with Mtphagy dye and Lyso dye, with mitophagy observed under a fluorescence microscope. The arrows show the co-localization of Mtphagy dye and Lyso dye, indicating the presence of mitophagy. (<b>B</b>) Western blot analysis revealed the protein levels of Tom20 and LC3I/II in HCC cells treated with TST (4 μM) for the indicated time point. Graphic representation of CI plot (Fa-CI plot) obtained from the CompuSyn software (version 1.0): (<b>C</b>) TST (2 μM, 4 μM) in combination with CQ (2, 4, 6, and 8 μM) for 24 h; (<b>D</b>) TST (2 μM, 4 μM) in combination with BafA1 (100, 200, 400, and 800 nM) for 24 h; (<b>E</b>) TST (2 μM, 4 μM) in combination with 3-MA (2, 4, 6, and 8 mM) for 24 h. CI &lt; 1 indicates synergistic, CI = 1 indicates additive, and CI &gt; 1 indicates antagonistic. 3-MA, 3-methyladenine; BafA1, bafilomycin A1; CQ, chloroquine.</p>
Full article ">
22 pages, 9316 KiB  
Article
The Role of Macrophage Polarization-Associated Gene Expression in the Oncological Prognosis of Hepatocellular Carcinoma
by Dong Liu, Yankun Li, Guanwu Wang, Edgar Dahl, Tom Luedde, Ulf Peter Neumann and Jan Bednarsch
Gastroenterol. Insights 2024, 15(3), 764-785; https://doi.org/10.3390/gastroent15030055 - 6 Sep 2024
Viewed by 477
Abstract
Background: The induced repolarization of tumor growth-promoting M2 macrophages into tumor growth-inhibiting M1 macrophages is a matter of intensive research and is expected to lead towards a novel targetable approach in HCC therapy. Methods: Differentially expressed M2 macrophage-related genes between normal and tumor [...] Read more.
Background: The induced repolarization of tumor growth-promoting M2 macrophages into tumor growth-inhibiting M1 macrophages is a matter of intensive research and is expected to lead towards a novel targetable approach in HCC therapy. Methods: Differentially expressed M2 macrophage-related genes between normal and tumor samples with high and low M2 macrophage infiltration in the Gene Expression Omnibus (GEO) and TCGA datasets were identified. A risk score was constructed based on univariate Cox analysis and LASSO-penalized Cox regression analysis. The relationship between the different risk score groups and clinical pathological characteristics as well as immune infiltration characteristics was studied. Subsequently, a nomogram was constructed to predict patients’ prognosis. Western blot and RT-qPCR were carried out to validate the results in human HCC samples. Results: Increased M2 macrophage infiltration was associated with a shorter overall survival (OS). Four important M2 macrophage-related genes (SLC22A1, CPS1, SLC10A1, CYP2C9) were discovered to be strongly correlated with OS and M2 macrophage infiltration. A nomogram incorporating the signature and tumor stage was developed for final clinical translation. Conclusions: SLC22A1, CPS1, SLC10A1 and CYP2C9 genes are associated with tumor-promoting M2 macrophage infiltration and might be potential targets for macrophage-related immunotherapy in HCC patients. Further, this four-gene signature is a potential tool for predicting prognosis in these patients. Full article
(This article belongs to the Section Liver)
Show Figures

Figure 1

Figure 1
<p>The immune infiltration landscape in HCC samples and survival analysis of M2 macrophages in TCGA cohort. (<b>A</b>) The generation, function and mechanisms of M1/M2 macrophages (<b>B</b>) The box plot of 22 types of immune cell infiltration abundances in cancer and normal groups. (<b>C</b>) The Kaplan–Meier survival analysis of the median value of M2 macrophages infiltration for HCC. (<b>D</b>) The optimal cutoff determined by the “survminer” package in R. (<b>E</b>) The Kaplan–Meier survival analysis of the optimal cut-off value of M2 macrophages infiltration for HCC. *** <span class="html-italic">p</span> &lt; 0.001, * <span class="html-italic">p</span> &lt; 0.05, ns not significant.</p>
Full article ">Figure 2
<p>Identification of DEGs and establishment of prognostic risk model. (<b>A</b>) Sixty differential expressed genes between HCC and normal tissues from eight public databases (<b>B</b>) The heatmap of differential expressed genes between high- and low-M2 macrophage infiltration groups in TCGA dataset. (<b>C</b>) The Venn plot of DEGs form (<b>A</b>,<b>B</b>). (<b>D</b>) The forest plot of prognostic M2 macrophage-related DEGs identified by univariate Cox analysis. (<b>E</b>) LASSO coefficient profiles of the 18 genes in the TCGA dataset. (<b>F</b>) Selection of the optimal parameter (lambda) in the LASSO model.</p>
Full article ">Figure 3
<p>Validation of the four-gene signature in the TCGA, ICGC, and GSE76427 datasets. (<b>A</b>–<b>C</b>) Risk-score distribution plots for the three datasets. In each plot, from top to bottom: distribution of riskscores (<b>A</b>), distribution of survival status (<b>B</b>), expression patterns of the four genes (<b>C</b>). (<b>D</b>–<b>F</b>) K–M survival curves showing OS of patients in the high- and low-risk-score subgroups across three datasets. (<b>G</b>–<b>I</b>) Time dependent ROC curves of the signature.</p>
Full article ">Figure 4
<p>Independent validation of the risk score for predicting overall survival of HCC. (<b>A</b>) Univariate and multivariate Cox analysis in TCGA cohort. (<b>B</b>–<b>D</b>) Riskscore distribution among T stage (<b>B</b>), tumor grading (<b>C</b>) and TNM stage (<b>D</b>). (<b>E</b>) Univariate and multivariate Cox analysis in ICGC cohort. (<b>F</b>) Univariate and multivariate Cox analysis in GSE76427 cohort. Bold <span class="html-italic">p</span> values indicate statistical significance.</p>
Full article ">Figure 5
<p>Development and validation of a nomogram predicting OS for HCC in TCGA cohort. (<b>A</b>) Development a nomogram for predicting the probability of 1-, 3-, and 5-year OS for HCC patients. (<b>B</b>) Calibration plot of the nomogram for predicting the probability of OS at 3 and 5 years. (<b>C</b>) ROC curve analyses of the nomogram. (<b>D</b>) Decision curve analysis of the net clinical benefit of the nomogram (red line), the signature (brown line), and AJCC stage (green line) for predicting OS of patients in TCGA. (<b>E</b>) Validation of the nomogram by calibration plot, ROC and decision curve analysis in ICGC cohort.</p>
Full article ">Figure 6
<p>The immune cell infiltration and immune-related factors landscape in HCC patients with high and low risk. (<b>A</b>) Heatmap of the correlation between risk score and several immune cells based on algorithm. (<b>B</b>) Boxplot of immune cell infiltration abundance based on CIBERSORT with significant expression differences between high- and low-risk groups. (<b>C</b>) Heatmap of the correlation between risk score and several immune cells based on ssGSEA algorithm. (<b>D</b>) Boxplot of immune cell infiltration abundance based on ssGSEA with significant expression differences between high- and low-risk groups. (<b>E</b>) Heatmap of the relationship between risk score and immunoinhibitory genes. (<b>F</b>) Boxplot of immunoinhibitory-related genes with significant expression differences between high and low risk groups. (<b>G</b>) Heatmap of the relationship between risk score and immunostimulatory genes. (<b>H</b>) Boxplot of immunostimulatory-related genes with significant expression differences between high- and low-risk groups. **** <span class="html-italic">p</span> &lt; 0:0001, *** <span class="html-italic">p</span> &lt; 0:001, ** <span class="html-italic">p</span> &lt; 0:01, * <span class="html-italic">p</span> &lt; 0:05.</p>
Full article ">Figure 7
<p>WGCNA analysis on DEGs and identification of module genes associated with immune score. (<b>A</b>) Analysis of network topology for soft powers. (<b>B</b>) Gene dendrogram and module colors. (<b>C</b>) Relationship between red module genes and immune score. (<b>D</b>) PPI network of hub genes. (<b>E</b>) Correlation between hub genes. (<b>F</b>) Correlation between hub genes and results of ESTIMATE. (<b>G</b>) GO enrichment of hub genes. (<b>H</b>) KEGG enrichment of hub genes.</p>
Full article ">
31 pages, 1385 KiB  
Review
Predictive Biomarkers and Resistance Mechanisms of Checkpoint Inhibitors in Malignant Solid Tumors
by Luciana Alexandra Pavelescu, Robert Mihai Enache, Oana Alexandra Roşu, Monica Profir, Sanda Maria Creţoiu and Bogdan Severus Gaspar
Int. J. Mol. Sci. 2024, 25(17), 9659; https://doi.org/10.3390/ijms25179659 - 6 Sep 2024
Viewed by 805
Abstract
Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor [...] Read more.
Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), defective deoxyribonucleic acid (DNA) mismatch repair (dMMR), microsatellite instability (MSI), and the tumor microenvironment (TME). Over the past decade, several types of ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, anti-programmed cell death 1 (PD-1) antibodies, anti-programmed cell death ligand 1 (PD-L1) antibodies, and anti-lymphocyte activation gene-3 (LAG-3) antibodies have been studied and approved by the Food and Drug Administration (FDA), with ongoing research on others. Recent studies highlight the critical role of the gut microbiome in influencing a positive therapeutic response to ICIs, emphasizing the importance of modeling factors that can maintain a healthy microbiome. However, resistance mechanisms can emerge, such as increased expression of alternative immune checkpoints, T-cell immunoglobulin (Ig), mucin domain-containing protein 3 (TIM-3), LAG-3, impaired antigen presentation, and alterations in the TME. This review aims to synthesize the data regarding the interactions between microbiota and immunotherapy (IT). Understanding these mechanisms is essential for optimizing ICI therapy and developing effective combination strategies. Full article
Show Figures

Figure 1

Figure 1
<p>Inhibition of PD-L1/PD-1 checkpoint. (<b>A</b>)<b>. T-cell-Inhibited Activation.</b> T Cell: This immune cell is essential for locating and eliminating cancer cells. Tumor Cell: A malignant cell that uses inhibitory pathways to avoid detection by the immune system. MHC-TCR Complex: The tumor cell’s major histocompatibility complex (MHC) binds an antigen to the TCR. For T cells to identify the tumor cell, this contact is necessary. When activated, a T-cell receptor known as PD-1 transmits an inhibitory signal that lessens the T cell’s capacity to assault the tumor. PD-L1 is a ligand expressed on the surface of tumor cells and attaches itself to T-cell PD-1. By blocking T-cell activation, this interaction enables the tumor cell to avoid immune recognition and elimination. Since the tumor cell, in this instance, expresses PD-L1, which binds to the T cell’s PD-1 receptor, the tumor cell’s capacity to attack the T cell is hindered. The T cell receives a “stop” signal from this contact, which stops it from destroying the tumor cell. (<b>B</b>)<b>. ICI Blockade.</b> T cells that have been fully activated can mount an immunological response. Anti-PD-1/PD-L1 Antibodies: These antibodies prevent the interaction between the tumor cell’s PD-L1 and the T cell’s PD-1. The inhibitory signal is stopped by obstructing this contact, which keeps the T cell activated. Tumor Cell Death and Immune Attack: As a result of the antibodies blocking the inhibitory signals, the T cell releases cytokines and other cytotoxic chemicals, which ultimately cause the tumor cell to be destroyed. Anti-PD-1 or anti-PD-L1 antibodies, which are ICIs, in this case, block the interaction that would typically inhibit the T cell. As a result, the tumor cell is successfully attacked and killed by the T cell, which still functions. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 18 August 2024).</p>
Full article ">Figure 2
<p>CTLA-4 checkpoint inhibition mechanism. Dendritic cell (DC): The APC type plays a crucial role in activating naive T cells. MHC: The MHC on the DC presents an antigen to the TCR on the naive T cell. This interaction is essential for T-cell activation. B7 molecules (CD80/CD86): These molecules on the DC surface bind to receptors on the T cell to either provide activation signals or engage in inhibitory signaling. Naive T cell: This T cell has not yet encountered its specific antigen. TCR: This receptor recognizes the antigen the MHC presents on the DC, initiating activation. CD28: A costimulatory receptor on the T cell that binds to B7 molecules (CD80/CD86) on the DC. When CD28 binds to B7, it provides a necessary costimulatory signal for T-cell activation, enhancing its response. CTLA-4: This receptor is also on the T cell and competes with CD28 for binding to B7. However, when CTLA-4 binds to B7, it sends an inhibitory signal, dampening the T cell’s activation to prevent excessive immune responses. Anti-CTLA-4 antibody: The figure shows an anti-CTLA-4 antibody blocking the CTLA-4 receptor. This prevents CTLA-4 from binding to B7 molecules, thereby blocking the inhibitory signal normally downregulating T-cell activation. By blocking CTLA-4, the antibody enhances the activation signal from the CD28-B7 interaction, promoting a stronger immune response. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 10 August 2024).</p>
Full article ">Figure 3
<p>The influence of intestinal microbiota in ICI treatment. DCs in the gut microbiome identify non-self-material and present it to T cells in the lymph nodes, activating memory T cells (TCMs). These TCMs differentiate into effector T cells and proliferate, increasing levels of human IL-2 and IFN-γ. Through their primary mechanisms, IL-2 and IFN-γ enhance the effectiveness of the therapeutic response to ICIs. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 10 August 2024).</p>
Full article ">
Back to TopTop