Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (239)

Search Parameters:
Keywords = resistance to anti-angiogenics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 1249 KiB  
Review
Bridging Immune Evasion and Vascular Dynamics for Novel Therapeutic Frontiers in Hepatocellular Carcinoma
by Sulin Wu, Namrata Anand, Zhoubo Guo, Mingyang Li, Marcos Santiago Figueroa, Lauren Jung, Sarah Kelly and Joseph W. Franses
Cancers 2025, 17(11), 1860; https://doi.org/10.3390/cancers17111860 - 31 May 2025
Viewed by 854
Abstract
Hepatocellular carcinoma (HCC) remains one of the most lethal cancers globally, driven by chronic liver disease and a complex tumor microenvironment (TME). Recent advances in spatial omics, single-cell analyses, and AI-driven digital pathology have shed light on the intricate heterogeneity of HCC, highlighting [...] Read more.
Hepatocellular carcinoma (HCC) remains one of the most lethal cancers globally, driven by chronic liver disease and a complex tumor microenvironment (TME). Recent advances in spatial omics, single-cell analyses, and AI-driven digital pathology have shed light on the intricate heterogeneity of HCC, highlighting key roles for immune suppression, angiogenesis, and fibrosis in tumor progression. This review synthesizes current epidemiological trends, noting a shift from viral hepatitis to metabolic syndrome as a primary etiology in Western populations, and elucidates how TME components—such as tumor-associated macrophages, cancer-associated fibroblasts, vascular endothelial cells, and immunosuppressive cytokines—contribute to resistance against conventional therapies. We detail the evolution of immunotherapeutic strategies from monotherapy to combination regimens, including dual immune checkpoint blockade and the integration of antiangiogenic agents. Emerging circulating and tissue-based biomarkers offer promise for enhanced patient stratification and real-time monitoring of treatment responses. Collectively, these innovations herald a paradigm shift toward TME-directed precision oncology in HCC, emphasizing the need for multi-targeted approaches to synergistically modulate interacting cellular constituents and ultimately improve clinical outcomes. Full article
(This article belongs to the Special Issue The Tumor Microenvironment: Interplay Between Immune Cells)
Show Figures

Figure 1

3 pages, 481 KiB  
Interesting Images
Tumor Cavitation with Anlotinib Treatment in Lung Adenocarcinoma
by Jie Huang and Xueqin Chen
Diagnostics 2025, 15(10), 1280; https://doi.org/10.3390/diagnostics15101280 - 18 May 2025
Viewed by 584
Abstract
Tumor cavitation is distinguished by the emergence of central necrosis and cavity formation within the tumor mass, which indicates a notable outcome of anti-angiogenic therapies. This case describes a 52-year-old Chinese female with advanced EGFR-mutated lung adenocarcinoma (Exon 19 deletion), which was metastatic [...] Read more.
Tumor cavitation is distinguished by the emergence of central necrosis and cavity formation within the tumor mass, which indicates a notable outcome of anti-angiogenic therapies. This case describes a 52-year-old Chinese female with advanced EGFR-mutated lung adenocarcinoma (Exon 19 deletion), which was metastatic to bilateral lungs, brain, and right adrenal gland, who exhibited a radiographic response to combination therapy with the third-generation EGFR tyrosine kinase inhibitor (TKI) aumolertinib and the anti-angiogenic agent anlotinib. The patient achieved near-complete cavitation of almost all bilateral lung nodules, manifesting as distinctive “bullet hole” lesions on the chest CT. Despite this initial response, disease progression occurred two months later with new liver metastases, culminating in the patient’s death. This case underscores the potential efficacy of EGFR TKIs and anti-angiogenic agents in inducing unique tumor microenvironment modifications, while highlighting the transient nature of such responses and the critical need to address resistance mechanisms. Tumor cavitation may serve as a radiographic marker of anti-angiogenic activity but does not preclude metastatic spread, necessitating vigilant monitoring even in the setting of favorable imaging changes. Full article
(This article belongs to the Section Medical Imaging and Theranostics)
Show Figures

Figure 1

24 pages, 2444 KiB  
Review
The CXCL1-CXCR2 Axis as a Component of Therapy Resistance, a Source of Side Effects in Cancer Treatment, and a Therapeutic Target
by Jan Korbecki, Mateusz Bosiacki, Maciej Pilarczyk, Marcin Kot, Piotr Defort, Ireneusz Walaszek, Dariusz Chlubek and Irena Baranowska-Bosiacka
Cancers 2025, 17(10), 1674; https://doi.org/10.3390/cancers17101674 - 15 May 2025
Viewed by 1220
Abstract
CXCL1 (Gro-α, MGSA) is a chemokine functionally similar to CXCL8/IL-8, as both activate the same receptor, CXCR2. CXCL1 levels are frequently elevated in tumors compared to healthy tissue, where they play a key role in promoting cancer cell migration, angiogenesis, and neutrophil recruitment. [...] Read more.
CXCL1 (Gro-α, MGSA) is a chemokine functionally similar to CXCL8/IL-8, as both activate the same receptor, CXCR2. CXCL1 levels are frequently elevated in tumors compared to healthy tissue, where they play a key role in promoting cancer cell migration, angiogenesis, and neutrophil recruitment. While the involvement of CXCL1 in tumor progression is well established, its relevance to cancer therapy remains underexplored. This review examines the therapeutic potential of targeting CXCL1 and its receptor, CXCR2, in cancer treatment. It discusses anti-CXCL1 antibodies and CXCR2 antagonists, including AZD5069, SB225002, SCH-479833, navarixin/SCH-527123, ladarixin/DF2156A, and reparixin, as well as strategies to enhance CXCR2 expression in lymphocytes during adoptive cell therapy to improve immunotherapy outcomes. Particular attention is given to the role of CXCL1 in treatment resistance, including resistance to chemotherapy, radiotherapy, and anti-angiogenic therapy. Cancer therapies often upregulate CXCL1 expression, which in turn drives treatment resistance. Additionally, this review explores the contribution of CXCL1 to therapy-induced side effects, such as chemotherapy-induced metastasis, neuropathy, nephrotoxicity, diarrhea, and cardiotoxicity. CXCR2 inhibitors are well tolerated by patients in clinical trials. However, the limited number of studies evaluating these agents in combination with standard chemotherapy precludes any definitive conclusions. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

16 pages, 810 KiB  
Review
Bruton’s Tyrosine Kinase: A Double-Edged Sword in Cancer and Aging
by Zahraa Qusairy and Miran Rada
Kinases Phosphatases 2025, 3(2), 10; https://doi.org/10.3390/kinasesphosphatases3020010 - 7 May 2025
Viewed by 992
Abstract
Bruton’s tyrosine kinase (BTK) is a key signaling molecule involved in both hematological malignancies and solid tumors. In B-cell malignancies such as chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL), BTK mediates B-cell receptor signaling, promoting tumor survival and proliferation, leading to the [...] Read more.
Bruton’s tyrosine kinase (BTK) is a key signaling molecule involved in both hematological malignancies and solid tumors. In B-cell malignancies such as chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL), BTK mediates B-cell receptor signaling, promoting tumor survival and proliferation, leading to the development of BTK inhibitors like ibrutinib that improve patient outcomes. In solid tumors, BTK isoforms, particularly p65BTK, contribute to tumor growth and therapy resistance, with inhibition showing promise in cancers like colorectal, ovarian, and non-small cell lung cancer. BTK also influences the tumor microenvironment by modulating immune cells such as myeloid-derived suppressor cells and tumor-associated macrophages, aiding immune evasion. BTK inhibition can enhance anti-tumor immunity and reduce inflammation-driven tumor progression. Additionally, BTK contributes to tumor angiogenesis, with inhibitors like ibrutinib showing anti-angiogenic effects. Beyond cancer, BTK is linked to aging, where its modulation may reduce senescent cell accumulation and preserve cognitive function. This review explores BTK’s dual role, focusing on its oncogenic effects and potential impact on aging processes. We also discuss the use of BTK inhibitors in cancer treatment and their potential to address age-related concerns, providing a deeper understanding of BTK as a therapeutic target and mediator in the complex relationship between cancer and aging. Full article
Show Figures

Figure 1

14 pages, 730 KiB  
Review
The Potential Therapeutic Role of Beta-Caryophyllene as a Chemosensitizer and an Inhibitor of Angiogenesis in Cancer
by Emad A. Ahmed
Molecules 2025, 30(8), 1751; https://doi.org/10.3390/molecules30081751 - 14 Apr 2025
Cited by 2 | Viewed by 1953
Abstract
The natural, highly lipophilic bicyclic sesquiterpenes, Beta-Caryophyllene (BCP), was highlighted in several recent preclinical studies to enhance chemo-sensitization in chemo-resistant tumors and to efficiently inhibit angiogenesis and cancer cells’ ability to invade and metastasize. Previous studies have researched the reasons for the synergistic [...] Read more.
The natural, highly lipophilic bicyclic sesquiterpenes, Beta-Caryophyllene (BCP), was highlighted in several recent preclinical studies to enhance chemo-sensitization in chemo-resistant tumors and to efficiently inhibit angiogenesis and cancer cells’ ability to invade and metastasize. Previous studies have researched the reasons for the synergistic effect of Beta-Caryophyllene in combination therapy and its role as a chemosensitizer and an inhibitor of angiogenesis through investigating the involved mechanisms and signaling molecules. These include the lipophilic nature of BCP, the selective interaction of BCP with CB2, the binding affinity of BCP to the receptor binding sites at the angiogenic vascular endothelial growth factor, and the upstream effect on JAK1/STAT3 pathway and other signaling pathways. Herein, the BCP role in enhancing chemo-sensitization of chemo-resistant tumors and in inhibiting angiogenesis and cancer cells’ ability to invade and metastasize are highlighted. Beta-Caryophyllene appears to be a promising candidate in treating cancer when co-supplemented with drugs such as cisplatin, gemcitabine and sorafenib. Clinical trials are needed to validate the potential therapeutic effect of BCP as a co-supplementary drug in cancer therapy, helping to sensitize cancer response to drugs, modulating signaling pathways, and lowering the drugs’ doses besides working as anti-angiogenetic drug. Full article
(This article belongs to the Special Issue Anticancer Natural Products)
Show Figures

Figure 1

21 pages, 653 KiB  
Review
Artemisinin and Its Derivatives: Promising Therapeutic Agents for Age-Related Macular Degeneration
by Chun Liu, Xiaoqin Liu and Junguo Duan
Pharmaceuticals 2025, 18(4), 535; https://doi.org/10.3390/ph18040535 - 6 Apr 2025
Viewed by 889
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment and blindness in older adults. Its pathogenesis involves multiple factors, including aging, environmental influences, genetic predisposition, oxidative stress, metabolic dysfunction, and immune dysregulation. Currently, AMD treatment focuses primarily on wet AMD, managed [...] Read more.
Age-related macular degeneration (AMD) is a leading cause of visual impairment and blindness in older adults. Its pathogenesis involves multiple factors, including aging, environmental influences, genetic predisposition, oxidative stress, metabolic dysfunction, and immune dysregulation. Currently, AMD treatment focuses primarily on wet AMD, managed through repeated intravitreal injections of anti-vascular endothelial growth factor (VEGF) therapies. While anti-VEGF agents represent a major breakthrough in wet AMD care, repeated injections may lead to incomplete responses or resistance in some patients, and carry a risk of progressive fibrosis. Artemisinin (ART) and its derivatives, originally developed as antimalarial drugs, exhibit a broad spectrum of pleiotropic activities beyond their established use, including anti-inflammatory, anti-angiogenic, antioxidant, anti-fibrotic, mitochondrial regulatory, lipid metabolic, and immunosuppressive effects. These properties position ART as a promising therapeutic candidate for AMD. A growing interest in ART-based therapies for AMD has emerged in recent years, with numerous studies demonstrating their potential benefits. However, no comprehensive review has systematically summarized the specific roles of ART and its derivatives in AMD pathogenesis and treatment. This paper aims to fill the knowledge gap by synthesizing the therapeutic efficacy and molecular mechanisms of ART and its derivatives in AMD, thereby providing a foundation for future investigations. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

54 pages, 10552 KiB  
Review
Plant Alkaloids as Promising Anticancer Compounds with Blood–Brain Barrier Penetration in the Treatment of Glioblastoma: In Vitro and In Vivo Models
by Marcin Ożarowski, Tomasz M. Karpiński, Bogusław Czerny, Adam Kamiński and Agnieszka Seremak-Mrozikiewicz
Molecules 2025, 30(7), 1561; https://doi.org/10.3390/molecules30071561 - 31 Mar 2025
Viewed by 2469
Abstract
Glioblastoma (GBM) is one of the most invasive central nervous system tumors, with rising global incidence. Therapy resistance and poor prognosis highlight the urgent need for new anticancer drugs. Plant alkaloids, a largely unexplored yet promising class of compounds, have previously contributed to [...] Read more.
Glioblastoma (GBM) is one of the most invasive central nervous system tumors, with rising global incidence. Therapy resistance and poor prognosis highlight the urgent need for new anticancer drugs. Plant alkaloids, a largely unexplored yet promising class of compounds, have previously contributed to oncology treatments. While past reviews provided selective insights, this review aims to collectively compare data from the last decade on (1) plant alkaloid-based anticancer drugs, (2) alkaloid transport across the blood–brain barrier (BBB) in vitro and in vivo, (3) alkaloid mechanisms of action in glioblastoma models (in vitro, in vivo, ex vivo, and in silico), and (4) cytotoxicity and safety profiles. Additionally, innovative drug delivery systems (e.g., nanoparticles and liposomes) are discussed. Focusing on preclinical studies of single plant alkaloids, this review includes 22 botanical families and 28 alkaloids that demonstrated anti-GBM activity. Most alkaloids act in a concentration-dependent manner by (1) reducing glioma cell viability, (2) suppressing proliferation, (3) inhibiting migration and invasion, (4) inducing cell death, (5) downregulating Bcl-2 and key signaling pathways, (6) exhibiting antiangiogenic effects, (7) reducing tumor weight, and (8) improving survival rates. The toxic and adverse effect analysis suggests that alkaloids such as noscapine, lycorine, capsaicin, chelerythrine, caffeine, boldine, and colchicine show favorable therapeutic potential. However, tetrandrine, nitidine, harmine, harmaline, cyclopamine, cocaine, and brucine may pose greater risks than benefits. Piperine’s toxicity and berberine’s poor bioavailability suggest the need for novel drug formulations. Several alkaloids (kukoamine A, cyclovirobuxine D, α-solanine, oxymatrine, rutaecarpine, and evodiamine) require further pharmacological and toxicological evaluation. Overall, while plant alkaloids show promise in glioblastoma therapy, progress in assessing their BBB penetration remains limited. More comprehensive studies integrating glioma research and advanced drug delivery technologies are needed. Full article
(This article belongs to the Special Issue Anti-cancer Agents from Natural Products)
Show Figures

Figure 1

26 pages, 4310 KiB  
Review
Addressing Challenges in Targeted Therapy for Metastatic Colorectal Cancer
by Maria El Hage, Zhaoran Su and Michael Linnebacher
Cancers 2025, 17(7), 1098; https://doi.org/10.3390/cancers17071098 - 25 Mar 2025
Cited by 5 | Viewed by 1655
Abstract
This review article aims to address the challenges associated with targeted therapy for the treatment of metastatic colorectal cancer (mCRC). We will first provide an overview of approved targeted therapies for treating mCRC, which include antiangiogenic therapy, as well as inhibitors of EGFR, [...] Read more.
This review article aims to address the challenges associated with targeted therapy for the treatment of metastatic colorectal cancer (mCRC). We will first provide an overview of approved targeted therapies for treating mCRC, which include antiangiogenic therapy, as well as inhibitors of EGFR, BRAFV600E, HER2 inhibitors, and immune checkpoints. Second, we discuss the different mechanisms of primary resistance, including tumor heterogeneity, both as inter-patient and intra-patient heterogeneity, and mechanisms of secondary resistance which include: driver oncogene alterations, downstream or parallel bypass signaling, presence of co-dominant driver oncogenes, tumor lineage plasticity, and epithelial to mesenchymal transition. Resistance mechanisms towards the different drug classes targeting mCRC are discussed in detail. Strategies to overcome resistance primarily involve combination of therapies, although this approach is typically linked to increased drug toxicity, manifesting as on and off-target effects. Moreover, the cost and accessibility of targeted therapies pose significant challenges for diverse populations. Addressing these challenges necessitates further research efforts aimed at optimizing the use of targeted therapy in mCRC. Integration of genomic biomarkers, such as sequencing and liquid biopsy, into routine clinical practice holds promise in enhancing treatment outcomes. In conclusion, this comprehensive review underscores the complex challenges encountered in targeted therapy for mCRC. Full article
(This article belongs to the Section Cancer Metastasis)
Show Figures

Figure 1

22 pages, 3057 KiB  
Review
Sulfonamides a Promising Hit for Cancer Therapy Through VEGFR-2 Inhibition
by Eleftherios Charissopoulos and Eleni Pontiki
Biomedicines 2025, 13(4), 772; https://doi.org/10.3390/biomedicines13040772 - 21 Mar 2025
Viewed by 890
Abstract
Vascular endothelial growth factor receptor-2 (VEGFR-2), a tyrosine kinase receptor (TKR), plays a crucial role in angiogenesis and is overexpressed in most cancers. It is important for tumor angiogenesis, facilitating essential angiogenic cellular processes, such as promoting endothelial cell survival, proliferation, migration, and [...] Read more.
Vascular endothelial growth factor receptor-2 (VEGFR-2), a tyrosine kinase receptor (TKR), plays a crucial role in angiogenesis and is overexpressed in most cancers. It is important for tumor angiogenesis, facilitating essential angiogenic cellular processes, such as promoting endothelial cell survival, proliferation, migration, and vascular permeability. Consequently, VEGFR-2 has become one of the main targets for anti-angiogenic therapy, with its inhibition serving as a crucial strategy for developing new drugs to mitigate angiogenesis-dependent cancers. Small-molecule drugs targeting VEGFR-2, approved by the USFDA, are exhibiting the development of drug resistance during chemotherapy, with cardiac-related side effects being consistently reported. In conclusion, it is important to develop novel strategies to enhance the efficacy of VEGFR-2 inhibitors and eliminate their adverse effects. Multifunctional drugs that target multiple pathways present a promising strategy, enhancing efficacy while minimizing side effects. Sulfonamide derivatives are extensively used in medicinal chemistry and modern drug discovery due to their variety of pharmacological activities. The present review focuses on novel compounds endowed with potential VEGFR-2 inhibition, four of which additionally present carbonic anhydrase inhibitory activity. Full article
(This article belongs to the Special Issue Recent Advances in Drug Synthesis and Drug Discovery)
Show Figures

Figure 1

37 pages, 2353 KiB  
Review
Precision Medicine in High-Grade Serous Ovarian Cancer: Targeted Therapies and the Challenge of Chemoresistance
by Sara Polajžer and Katarina Černe
Int. J. Mol. Sci. 2025, 26(6), 2545; https://doi.org/10.3390/ijms26062545 - 12 Mar 2025
Cited by 3 | Viewed by 2014
Abstract
The poor prognosis for high-grade serous ovarian cancer (HGSOC), the dominant subtype of ovarian cancer, reflects its aggressive nature, late diagnosis, and the highest mortality rate among all gynaecologic cancers. Apart from late diagnosis, the main reason for the poor prognosis and its [...] Read more.
The poor prognosis for high-grade serous ovarian cancer (HGSOC), the dominant subtype of ovarian cancer, reflects its aggressive nature, late diagnosis, and the highest mortality rate among all gynaecologic cancers. Apart from late diagnosis, the main reason for the poor prognosis and its unsuccessful treatment is primarily the emergence of chemoresistance to carboplatin. Although there is a good response to primary treatment, the disease recurs in 80% of cases, at which point it is largely resistant to carboplatin. The introduction of novel targeted therapies in the second decade of the 21st century has begun to transform the treatment of HGSOC, although their impact on overall survival remains unsatisfactory. Targeting the specific pathways known to be abnormally activated in HGSOC is especially difficult due to the molecular diversity of its subtypes. Moreover, a range of molecular changes are associated with acquired chemoresistance, e.g., reversion of BRCA1 and BRCA2 germline alleles. In this review, we examine the advantages and disadvantages of approved targeted therapies, including bevacizumab, PARP inhibitors (PARPis), and treatments targeting cells with neurotrophic tyrosine receptor kinase (NTRK), B-rapidly accelerated fibrosarcoma (BRAF), and rearranged during transfection (RET) gene alterations, as well as antibody–drug conjugates. Additionally, we explore promising new targets under investigation in ongoing clinical trials, such as immune checkpoint inhibitors, anti-angiogenic agents, phosphatidylinositol-3-kinase (PI3K) inhibitors, Wee1 kinase inhibitors, and ataxia telangiectasia and Rad3-related protein (ATR) inhibitors for platinum-resistant disease. Despite the development of new targeted therapies, carboplatin remains the fundamental medicine in HGSOC therapy. The correct choice of treatment strategy for better survival of patients with advanced HGSOC should therefore include a prediction of patients’ risks of developing chemoresistance to platinum-based chemotherapy. Moreover, effective targeted therapy requires the selection of patients who are likely to derive clinical benefit while minimizing potential adverse effects, underscoring the essence of precision medicine. Full article
(This article belongs to the Special Issue Molecular Pathology and Treatment of Ovarian Cancer)
Show Figures

Figure 1

30 pages, 1360 KiB  
Review
Angiogenesis in Glioblastoma—Treatment Approaches
by Agnieszka Nowacka, Maciej Śniegocki, Wojciech Smuczyński, Dominika Bożiłow and Ewa Ziółkowska
Cells 2025, 14(6), 407; https://doi.org/10.3390/cells14060407 - 11 Mar 2025
Cited by 5 | Viewed by 1986
Abstract
Glioblastoma, the most common primary malignant brain tumor in adults, carries a poor prognosis, with a median survival of just 15 months, significantly impacting patients’ quality of life. The aggressive growth of these highly vascularized tumors relies heavily on angiogenesis, driven primarily by [...] Read more.
Glioblastoma, the most common primary malignant brain tumor in adults, carries a poor prognosis, with a median survival of just 15 months, significantly impacting patients’ quality of life. The aggressive growth of these highly vascularized tumors relies heavily on angiogenesis, driven primarily by vascular endothelial growth factor-A. Therefore, VEGF signaling pathway has become a prime therapeutic target in GBM treatment over the past decade. While anti-angiogenic treatment showed promise, agents like bevacizumab have ultimately failed to improve overall survival. This highlights the presence of compensatory angiogenic mechanisms that bypass VEGF inhibition, necessitating further investigation into resistance mechanisms and the development of more effective therapeutic strategies. This review examined the current landscape of anti-angiogenic agents for GBM, analyzed the mechanisms driving resistance to these therapies, and explored potential strategies for enhancing their effectiveness. Full article
(This article belongs to the Special Issue Therapeutic Targets in Glioblastoma)
Show Figures

Figure 1

25 pages, 4398 KiB  
Article
Influence of a Very High-Molecular Weight Fucoidan from Laminaria hyperborea on Age-Related Macular Degeneration-Relevant Pathomechanisms in Ocular Cell Models
by Philipp Dörschmann, Georg Kopplin, Tabea Thalenhorst, Charlotte Seeba, Sadia Fida Ullah, Vaibhav Srivastava, Johann Roider and Alexa Klettner
Mar. Drugs 2025, 23(3), 101; https://doi.org/10.3390/md23030101 - 25 Feb 2025
Viewed by 1093
Abstract
Fucoidans from Laminaria hyperborea (LH) can be antioxidative, antiangiogenic, and anti-inflammatory. In this study, a very high-molecular weight (3700 kDa) fucoidan from LH, FucBB04, was tested regarding its bioactivity in age-related macular degeneration (AMD) models in vitro. Primary retinal pigment epithelium (RPE) [...] Read more.
Fucoidans from Laminaria hyperborea (LH) can be antioxidative, antiangiogenic, and anti-inflammatory. In this study, a very high-molecular weight (3700 kDa) fucoidan from LH, FucBB04, was tested regarding its bioactivity in age-related macular degeneration (AMD) models in vitro. Primary retinal pigment epithelium (RPE) from pig eyes, human uveal melanoma cell line OMM-1, and RPE cell line ARPE-19 were used. Substituents of the extract were determined with chemical analysis. Cell viability was tested with tetrazolium assay (MTT), oxidative stress was induced by H2O2 or erastin, respectively. Secreted vascular endothelial growth factor A (VEGF-A) was assessed with ELISA. Retinal pigment epithelium 65 kDa protein (RPE65) and protectin (CD59) protein expression were tested in Western blot. Cell barrier was assessed by measuring trans-epithelial electrical resistance (TEER), phagocytic ability by a fluorescence assay. Gene expression and secretion of interleukin 6 (IL-6) and interleukin 8 (IL-8) were tested in real-time PCR and ELISA. FucBB04 displayed no oxidative stress protective effects. Its effect on VEGF was inconsistent, with VEGF secretion reduced in primary RPE, but not in ARPE-19. On the other hand, Lipopolysaccharide (LPS) and polyinosinic/polycytidylic acid (PIC)-induced IL-6 or IL-8 secretion was reduced by FucBB04, while complement inhibiting protein CD59 was not affected. In addition, FucBB04 did not influence the gene expression of IL-6 or IL-8. Visual cycle protein RPE65 expression, phagocytic ability, and barrier function were reduced by FucBB04. Very high-molecular weight fucoidan from LH shows bioactivities against AMD-related pathological pathways, but adverse effects on RPE function may limit its suitability as a therapeutic compound. Smaller high-molecular weight fucoidans are recommended for further research. Full article
(This article belongs to the Special Issue Marine Anti-Inflammatory and Antioxidant Agents, 4th Edition)
Show Figures

Figure 1

25 pages, 1216 KiB  
Review
Chondrosarcoma: New Molecular Insights, Challenges in Near-Patient Preclinical Modeling, and Therapeutic Approaches
by Lorena Landuzzi, Francesca Ruzzi, Pier-Luigi Lollini and Katia Scotlandi
Int. J. Mol. Sci. 2025, 26(4), 1542; https://doi.org/10.3390/ijms26041542 - 12 Feb 2025
Cited by 2 | Viewed by 2033
Abstract
Chondrosarcoma (CS), the second most common malignant bone tumor after osteosarcoma, accounts for 20–30% of all malignant bone tumors. It mainly affects adults, middle-aged, and elderly people. The CS family includes various entities displaying peculiar biological, genetic, and epigenetic characteristics and clinical behaviors. [...] Read more.
Chondrosarcoma (CS), the second most common malignant bone tumor after osteosarcoma, accounts for 20–30% of all malignant bone tumors. It mainly affects adults, middle-aged, and elderly people. The CS family includes various entities displaying peculiar biological, genetic, and epigenetic characteristics and clinical behaviors. Conventional CS is the most common subtype. High-grade, dedifferentiated, and mesenchymal CS, as well as unresectable and metastatic CS, exhibit poor prognoses due to their intrinsic resistance to radiotherapy and chemotherapy, underscoring the urgent need for novel therapeutic strategies. CS research is dealing with several challenges. Experimental studies can rely on animal and patient-derived models, but the paucity of representative near-patient preclinical models has hampered predictive drug screening research. This review describes the main clinical and molecular features of CS subtypes, discussing recent data on the genetic alterations and molecular mechanisms involved in CS pathogenesis and progression. The review provides an overview of the current in vitro and in vivo CS models, discusses their advantages and limitations, and highlights the recent efforts in the development of new targeted therapies against CS dependencies, including IDH1/2 mutations, NAD+ dependency, and SIRT1-HIF-2α axis, or exploring DR5 targeting, antiangiogenic therapies, epigenetic drugs, and immunological approaches. All such strategies, in combination with advanced preclinical modeling and personalized multi-omic profiling, hold promise for improving the survival of patients with advanced CS. Full article
(This article belongs to the Special Issue Molecular Research in Bone and Soft Tissue Tumors)
Show Figures

Figure 1

21 pages, 2703 KiB  
Article
Gallic Acid Enhances Olaparib-Induced Cell Death and Attenuates Olaparib Resistance in Human Osteosarcoma U2OS Cell Line
by Mehmet Kadir Erdogan and Ayse Busra Usca
Curr. Issues Mol. Biol. 2025, 47(2), 104; https://doi.org/10.3390/cimb47020104 - 7 Feb 2025
Viewed by 1317
Abstract
Cancer remains one of the most formidable diseases globally and continues to be a leading cause of mortality. While chemotherapeutic agents are crucial in cancer treatment, they often come with severe side effects. Furthermore, the development of acquired drug resistance poses a significant [...] Read more.
Cancer remains one of the most formidable diseases globally and continues to be a leading cause of mortality. While chemotherapeutic agents are crucial in cancer treatment, they often come with severe side effects. Furthermore, the development of acquired drug resistance poses a significant challenge in the ongoing battle against cancer. Combining these chemotherapeutic agents with plant-derived phenolic compounds offers a promising approach, potentially reducing side effects and counteracting drug resistance. Phytochemicals, the bioactive compounds found in plants, exhibit a range of health-promoting properties, including anticarcinogenic, antimutagenic, antiproliferative, antioxidant, antimicrobial, neuroprotective, and cardioprotective effects. Their ability to enhance treatment, coupled with their non-toxic, multi-targeted nature and synergistic potential when used alongside conventional drugs, underscores the growing importance of natural therapeutics. In this study, we investigated the anticancer effects of olaparib (OL), a small-molecule PARP inhibitor that has shown promising results in both preclinical and clinical trials, and gallic acid (GA), a phenolic compound, in olaparib-resistant human osteosarcoma U2OS cells (U2OS-PIR). Both parental U2OS and U2OS-PIR cell lines were treated with increasing concentrations of olaparib and gallic acid, and their cytotoxic effects were assessed using the WST-1 cell viability assay. The synergistic potential of OL and GA, based on their determined IC50 values, was further explored in combination treatment. A colony survival assay revealed the combination’s ability to significantly reduce the colony-forming capacity of cancer cells. Additionally, the apoptotic effects of OL and GA, both individually and in combination, were examined in U2OS-PIR cells using acridine orange/ethidium bromide dual staining. The anti-angiogenic properties were assessed through a VEGF ELISA, while the expression of proteins involved in DNA damage and apoptotic signaling pathways was analyzed via Western blot. The results of this study demonstrate that gallic acid effectively suppresses cell viability and colony formation, particularly when used in combination therapy to combat OL resistance. Additionally, GA inhibits angiogenesis and induces DNA damage and apoptosis by modulating key apoptosis-related proteins, including cPARP, Bcl-2, and Bax. These findings highlight gallic acid as a potential compound for enhancing therapeutic efficacy in overcoming acquired drug resistance. Full article
(This article belongs to the Special Issue The Role of Natural Compounds in Cancer Therapy)
Show Figures

Figure 1

35 pages, 1042 KiB  
Review
Optimizing Osimertinib for NSCLC: Targeting Resistance and Exploring Combination Therapeutics
by Yan-You Liao, Chia-Luen Tsai and Hsiang-Po Huang
Cancers 2025, 17(3), 459; https://doi.org/10.3390/cancers17030459 - 29 Jan 2025
Cited by 2 | Viewed by 4886
Abstract
Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, with epidermal growth factor receptor (EGFR) mutations present in a substantial proportion of patients. Third-generation EGFR tyrosine kinase inhibitors (EGFR TKI), exemplified by osimertinib, have dramatically improved outcomes by effectively targeting [...] Read more.
Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, with epidermal growth factor receptor (EGFR) mutations present in a substantial proportion of patients. Third-generation EGFR tyrosine kinase inhibitors (EGFR TKI), exemplified by osimertinib, have dramatically improved outcomes by effectively targeting the T790M mutation—a primary driver of acquired resistance to earlier-generation EGFR TKI. Despite these successes, resistance to third-generation EGFR TKIs inevitably emerges. Mechanisms include on-target mutations such as C797S, activation of alternative pathways like MET amplification, histologic transformations, and intricate tumor microenvironment (TME) alterations. These resistance pathways are compounded by challenges in tolerability, adverse events, and tumor heterogeneity. In light of these hurdles, this review examines the evolving landscape of combination therapies designed to enhance or prolong the effectiveness of third-generation EGFR TKIs. We explore key strategies that pair osimertinib with radiotherapy, anti-angiogenic agents, immune checkpoint inhibitors, and other molecularly targeted drugs, and we discuss the biological rationale, preclinical evidence, and clinical trial data supporting these approaches. Emphasis is placed on how these combinations may circumvent diverse resistance mechanisms, improve survival, and maintain a favorable safety profile. By integrating the latest findings, this review aims to guide clinicians and researchers toward more individualized and durable treatment options, ultimately enhancing both survival and quality of life for patients with EGFR-mutated NSCLC. Full article
Show Figures

Figure 1

Back to TopTop