[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (6,303)

Search Parameters:
Keywords = antitumor activities

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 5649 KiB  
Review
The Significant Therapeutic Effects of Chinese Scorpion: Modern Scientific Exploration of Ion Channels
by Yueyuan Zheng, Qiuyi Wen, Yushi Huang and Dean Guo
Pharmaceuticals 2024, 17(12), 1735; https://doi.org/10.3390/ph17121735 (registering DOI) - 22 Dec 2024
Viewed by 102
Abstract
Chinese scorpion (CS), a traditional animal-based medicine used for over a millennium, has been documented since AD 935–960. It is derived from the scorpion Buthus martensii Karsch and is used to treat various ailments such as stroke, epilepsy, rheumatism, and more. Modern research [...] Read more.
Chinese scorpion (CS), a traditional animal-based medicine used for over a millennium, has been documented since AD 935–960. It is derived from the scorpion Buthus martensii Karsch and is used to treat various ailments such as stroke, epilepsy, rheumatism, and more. Modern research has identified the pharmacological mechanisms behind its traditional uses, with active components like venom and proteins showing analgesic, antitumor, antiepileptic, and antithrombotic effects. Studies reveal that CS affects ion channels, crucial for cellular functions, through interactions with sodium, potassium, and calcium channels, potentially explaining its therapeutic effects. Future research aims to elucidate the precise mechanisms, target specific ion channel subtypes, and validate clinical efficacy and safety, paving the way for novel therapies based on these natural compounds. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

Figure 1
<p>The interaction of scorpion toxins with specific ion channels elicits antitumor effects. (<b>A</b>) Modulation of cellular targets involved in cancer cell proliferation and apoptosis mechanisms by sodium channels scorpion toxins. (<b>B</b>) Inhibition of cellular targets involved in cancer cell proliferation and migration mechanisms by potassium channels scorpion toxins. (<b>C</b>) Inhibition of cellular targets involved in cancer cell invasion and migration mechanisms by chloride channel scorpion toxins [<a href="#B12-pharmaceuticals-17-01735" class="html-bibr">12</a>] (reproduced with permission from Najet Srairi-Abid, Cell Calcium; published by Elsevier, 2019).</p>
Full article ">Figure 2
<p>Mode of action for peptides with analgesic activity [<a href="#B22-pharmaceuticals-17-01735" class="html-bibr">22</a>] (reproduced with permission from Zhongjie Li, Peptides; published by Elsevier, 2019).</p>
Full article ">Figure 3
<p>Diagram illustrating the pathways associated with AGAP. (<b>A</b>) AGAP induces analgesia by effectively alleviating acute inflammatory pain and chronic constrictive injury through the modulation of MAPK and Fos signaling pathways in formalin-induced models. (<b>B</b>) AGAP decreases breast cancer cell stemness and epithelial–mesenchymal transition. (<b>C</b>) AGAP influences SHG-44 human malignant glioma cells and colon cancer cells [<a href="#B24-pharmaceuticals-17-01735" class="html-bibr">24</a>] (reproduced with permission from Seidu A. Richard, Evidence-Based Complementary and Alternative Medicine; published by Hindawi, 2020).</p>
Full article ">Figure 4
<p>The structure of potassium channels [<a href="#B40-pharmaceuticals-17-01735" class="html-bibr">40</a>] (reproduced with permission from Chenglai Xia, Biomedicine &amp; Pharmacotherapy; published by Elsevier, 2023).</p>
Full article ">Figure 5
<p>Proposed mechanism for the interaction between BK channels (<span class="html-italic">α</span> + <span class="html-italic">β</span>4) and martentoxin. The conformation of a normal open state (<b>A</b>) was changed by the application of martentoxin when the first steady complex was formed (<b>B</b>). Then, the low-affinity site was exposed to associate with martentoxin (<b>C</b>) or iberiotoxin (<b>D</b>) [<a href="#B62-pharmaceuticals-17-01735" class="html-bibr">62</a>] (reproduced with permission from Yonghua Ji, Biophysical Journal; published by Elsevier, 2008).</p>
Full article ">Figure 6
<p>Mode of action of martentoxin and bumarsin. KCa: Ca<sup>2+</sup>-activated K<sup>+</sup> channels; iNOS: inducible nitric oxide synthase; NO: nitric oxide [<a href="#B22-pharmaceuticals-17-01735" class="html-bibr">22</a>] (reproduced with permission from Zhongjie Li, Peptides; published by Elsevier, 2019).</p>
Full article ">Figure 7
<p>Structures of special small-molecule compounds in CS.</p>
Full article ">Figure 8
<p>Sequence alignment of the 22 identified full-length scorpion toxins. Similarities in sequences are indicated with shading. The six conserved cysteine residues in all toxins, capable of forming three disulfide bonds, are illustrated by the red box with connecting lines [<a href="#B92-pharmaceuticals-17-01735" class="html-bibr">92</a>] (reproduced with permission from Yingliang Wu, Journal of Proteomics; published by Elsevier, 2019).</p>
Full article ">
23 pages, 11843 KiB  
Article
C118P Suppresses Gastric Cancer Growth via Promoting Autophagy–Lysosomal Degradation of RAB1A
by Shihui Wei, Jing Zhang, Hai Wu, Zhengguang Liao, Zhengrui Liu, Yuhang Hou, Danyu Du, Jingwei Jiang, Li Sun, Shengtao Yuan and Mei Yang
Pharmaceutics 2024, 16(12), 1620; https://doi.org/10.3390/pharmaceutics16121620 (registering DOI) - 21 Dec 2024
Viewed by 314
Abstract
Background/Objectives: Gastric cancer (GC) is the leading cause of cancer-related deaths worldwide. C118P, a microtubule inhibitor with anti-angiogenic and vascular-disrupting activities, was proven to be cytotoxic to various cancer cell lines. This study aimed to explore the anti-tumor effect of C118P against [...] Read more.
Background/Objectives: Gastric cancer (GC) is the leading cause of cancer-related deaths worldwide. C118P, a microtubule inhibitor with anti-angiogenic and vascular-disrupting activities, was proven to be cytotoxic to various cancer cell lines. This study aimed to explore the anti-tumor effect of C118P against gastric cancer and identify its potential target. Methods: The MTT assay, colony formation assay, and EdU incorporation assay were used to evaluate the effect of C118P on GC cell proliferation. Cell cycle and cell apoptosis were measured using flow cytometry. Molecular docking, a microscale thermophoresis (MST) analysis, and the cellular thermal shift assay (CETSA) were used to investigate the binding of C118P to RAB1A. Autophagy-related effects were evaluated by using the MDC staining assay, immunofluorescence assay, and immunoblotting assay. The SGC-7901 cell line xenograft mouse model was used to confirm the anti-tumor efficacy of C118P. Results: C118P dramatically inhibited proliferation, induced G2/M cell cycle arrest, and triggered apoptosis in GC cell lines HGC-27 and SGC-7901. Mechanistically, C118P was demonstrated to bind with RAB1A and reduce the RAB1A protein level, accompanied by the inhibition of mTORC1 signaling. Moreover, C118P induced autophagosome formation and promoted RAB1A protein degradation in an autophagy–lysosomal-dependent manner. The in vivo study verified that C118P inhibits GC growth by inhibiting the RAB1A-mTOR axis. Conclusions: Our findings suggested that C118P inhibits GC growth by promoting the autophagy–lysosomal-dependent degradation of RAB1A and modulating mTOR C1 signaling. C118P shows potential as being a small molecule drug effective in the treatment of gastric cancer via targeting RAB1A. Full article
(This article belongs to the Section Drug Targeting and Design)
23 pages, 10599 KiB  
Article
Microbubble-Protected Oncolytic Virotherapy Targeted by Sonoporation Induces Tumor Necrosis and T-Lymphocyte Infiltration in Humanized Mice Bearing Triple-Negative Breast Cancer
by Juliana Sitta, Flavia De Carlo, Imani Kirven, John H. Tackett, Patrice Penfornis, George Clement Dobbins, Mallory Barbier, Luis Del Valle, Clayton T. Larsen, Ernest G. Schutt, Rhodemann Li, Candace M. Howard and Pier Paolo Claudio
Int. J. Mol. Sci. 2024, 25(24), 13697; https://doi.org/10.3390/ijms252413697 (registering DOI) - 21 Dec 2024
Viewed by 310
Abstract
Oncolytic virotherapy has shown great promise in mediating targeted tumor destruction through tumor-selective replication and induction of anti-tumor immunity; however, obstacles remain for virus candidates to reach the clinic. These include avoiding neutralizing antibodies, preventing stimulation of the adaptive immune response during intravenous [...] Read more.
Oncolytic virotherapy has shown great promise in mediating targeted tumor destruction through tumor-selective replication and induction of anti-tumor immunity; however, obstacles remain for virus candidates to reach the clinic. These include avoiding neutralizing antibodies, preventing stimulation of the adaptive immune response during intravenous administration, and inducing sufficient apoptosis and immune activation so that the body’s defense can work to eradicate systemic disease. We have developed a co-formulation of oncolytic viruses (OVs) with Imagent® lipid-encapsulated, perfluorocarbon microbubbles (MBs) to protect the OVs from the innate and adaptive immune system. Once inside the MB, the viral particles become acoustically active such that external ultrasound can target the delivery of the virus locally within the tumor. Humanized NSG female mice (Hu-CD34+ NSG-SGM3) engrafted in their flanks with MDA-MB-231-Luc triple-negative breast cancer (TNBC) cells were transduced with MB/OVs, with or without adjuvant Pembrolizumab treatment, and tumor sizes and tumor necrosis were assessed. The presence of CD8+ (cytotoxic T-cells), CD4+ (helper T-cells), and CD25+ (Tregs) tumor-infiltrating lymphocytes (TILs) was quantified in the tumor samples by immunohistochemistry. In an in vivo model of humanized mice engrafted with a human immune system, we observed significantly greater tumor necrosis and smaller tumor mass in human TNBC xenografts systemically treated with MB/OV complexes in the presence or absence of pembrolizumab adjuvant treatment, compared to controls. Additionally, we observed a low ratio of CD4+/CD8+ TILs and a high ratio of CD8+/CD25+ TILs in the MDA-MB-231 xenografts treated with MB/OVs complexes with or without pembrolizumab adjuvant treatment, compared to controls. Our study demonstrated the feasibility of using MBs to target OVs to TNBC through diagnostic ultrasound, which decreased tumor mass by increasing tumor necrosis and stimulated a local and systemic antitumoral immune response by increasing intratumoral CD8+ T-cytotoxic lymphocyte infiltration and decreasing CD25+ Treg cells. Full article
(This article belongs to the Special Issue Molecular Research in Triple-Negative Breast Cancer)
Show Figures

Figure 1

Figure 1
<p>Study treatment schedule. Treatments started when tumors were 150 mm<sup>3</sup>. Created with <a href="http://Biorender.com" target="_blank">Biorender.com</a>.</p>
Full article ">Figure 2
<p>Detection of Adenovirus-5 immunohistochemistry. Adenovirus-5 Hexon protein expression was negative in the controls (MBs and Pembrolizumab groups). Adenovirus-5 Hexon expression significantly increased in the OV group, with a 50–75% rate in infected cells in the IV groups. The magnification of the upper panels is 100×, and the lower panels is 200×.</p>
Full article ">Figure 3
<p>Tumor measurements by caliper and by IVIS.</p>
Full article ">Figure 4
<p>Hematoxylin and eosin staining of tumor xenografts (right flank). A montage of the treated tumor is shown on the upper panels, where the areas of necrosis are evident; lower panels show a low magnification view of the tumors in which the different sizes of the necrotic areas can be observed (original magnification 100×).</p>
Full article ">Figure 5
<p>Detection of tumor-infiltrating CD4<sup>+</sup> cells by immunohistochemistry. CD4-positive immune cells are few in the control (saline) group and sporadic in the Pembrolizumab-treated group, but their numbers significantly increase in the OV groups and dramatically increase in the MB groups. The upper panel magnification is 100×, and the lower panel magnification is 200×.</p>
Full article ">Figure 6
<p>Immunohistochemical detection of CD8<sup>+</sup> T-cells in tumors. CD8<sup>+</sup> T cell tumor infiltrates (TILs) are few in the control (saline) group, and sporadic in the Pembrolizumab treated group; however, their expression significantly increases in both OV groups, and their number is dramatically increased in both MB groups. The magnification of the upper panels is 100×, and the magnification of the lower panels is 200×.</p>
Full article ">Figure 7
<p>Immunohistochemistry for IL-R2 alpha (CD25) in tumors. CD25<sup>+</sup> cells are regularly distributed in the control MB group and are abundant in the Pembrolizumab group, mainly located within or surrounding areas of necrosis; however, their number plummeted in both MB groups, coinciding with these tumors having significantly smaller areas of necrosis. The magnification of the upper panels is 100×, and the magnification of the lower panels is 200×.</p>
Full article ">
31 pages, 1122 KiB  
Review
Therapeutic Significance of NLRP3 Inflammasome in Cancer: Friend or Foe?
by Aliea M. Jalali, Kenyon J. Mitchell, Christian Pompoco, Sudeep Poludasu, Sabrina Tran and Kota V. Ramana
Int. J. Mol. Sci. 2024, 25(24), 13689; https://doi.org/10.3390/ijms252413689 (registering DOI) - 21 Dec 2024
Viewed by 387
Abstract
Besides various infectious and inflammatory complications, recent studies also indicated the significance of NLRP3 inflammasome in cancer progression and therapy. NLRP3-mediated immune response and pyroptosis could be helpful or harmful in the progression of cancer, and also depend on the nature of the [...] Read more.
Besides various infectious and inflammatory complications, recent studies also indicated the significance of NLRP3 inflammasome in cancer progression and therapy. NLRP3-mediated immune response and pyroptosis could be helpful or harmful in the progression of cancer, and also depend on the nature of the tumor microenvironment. The activation of NLRP3 inflammasome could increase immune surveillance and the efficacy of immunotherapy. It can also lead to the removal of tumor cells by the recruitment of phagocytic macrophages, T-lymphocytes, and other immune cells to the tumor site. On the other hand, NLRP3 activation can also be harmful, as chronic inflammation driven by NLRP3 supports tumor progression by creating an environment that facilitates cancer cell proliferation, migration, invasion, and metastasis. The release of pro-inflammatory cytokines such as IL-1β and IL-18 can promote tumor growth and angiogenesis, while sustained inflammation may lead to immune suppression, hindering effective anti-tumor responses. In this review article, we discuss the role of NLRP3 inflammasome-mediated inflammatory response in the pathophysiology of various cancer types; understanding this role is essential for the development of innovative therapeutic strategies for cancer growth and spread. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Canonical and non-canonical activation of NLRP3 inflammasome. The canonical pathway involves the activation of NLRP3 inflammasomes through signals such as mitochondrial ROS, calcium influx, and potassium efflux, leading to NF-κB activation and the production of pro-inflammatory cytokines (IL-1β and IL-18). This pathway ultimately activates caspase-1, resulting in cytokine release and pyroptosis. The non-canonical pathway involving LPS from Gram-negative bacteria triggers caspase-11, which indirectly activates NLRP3, leading to similar inflammasome responses, cytokine production, and pyroptosis. Both pathways generally play critical roles in innate immunity and inflammation.</p>
Full article ">Figure 2
<p>Significance of NLRP3 inflammasome in the melanoma progression. Various factors such as UV radiation, tumor microenvironment, and melanoma cells contribute to oxidative stress and cause DNA damage, immune cell activation, and cytokine release. Oxidative stress, in turn, triggers reactive oxygen species (ROS), mitochondrial DNA damage, potassium efflux, and NF-κB activation, which influence the activation of the NLRP3 inflammasome. NLRP3 activation promotes melanoma progression, metastasis, immune evasion, and therapy resistance. In contrast, inhibition of NLRP3 could enhance immunotherapy, inhibit tumor growth, and reduce metastasis.</p>
Full article ">Figure 3
<p>Significance of NLRP3 inflammasome in leukemias. Oxidative stress and mitochondrial dysfunction in leukemia cells could activate NLRP3 inflammasomes through mitochondrial ROS, potassium efflux, and NF-κB signaling pathways. NLRP3 activation leads to the generation of active IL-1β and IL-18 cytokines, and could cause pyroptosis. Further, NLRP3 activation plays various roles in different leukemias. For example, in Acute Myeloid Leukemia (AML), NLRP3 promotes immune evasion and survival, while inhibition reduces the disease burden. In Chronic Myeloid Leukemia (CML), NLRP3 is linked to KRAS mutations and therapy resistance. In Acute Lymphoblastic Leukemia (ALL), NLRP3 activation is correlated with glucocorticoid resistance, and in Chronic Lymphocytic Leukemia (CLL), P2X7R overexpression leads to increased NLRP3.</p>
Full article ">Figure 4
<p>Role of NLRP3 inflammasome in breast cancer growth and spread. Several factors such as reactive oxygen species (ROS)-induced mitochondrial damage, BRCA1-associated genetic mutations causing mitochondrial dysfunction, extracellular ATP leading to P2X7R overexpression, and inflammatory cytokines that activate NF-κB-mediated inflammasome components could lead to activation of NLRP3 inflammasome. NLRP3-mediated release of IL-1β and IL-18 promotes cancer cell proliferation, survival, migration, immune evasion, and resistance to therapy. Further, the outcomes also include increased tumor growth, metastasis, compromised immune surveillance, and drug resistance.</p>
Full article ">Figure 5
<p>Significance of NLRP3 inflammasome activation in cigarette smoke and COPD-induced lung cancer development. Cigarette smoke leads to reactive oxygen species (ROS), mitochondrial damage, and tissue dysfunction, activating the NLRP3 inflammasome. The release of pro-inflammatory cytokines IL-1β and IL-18 contributes to prolonged inflammation. COPD-induced inflammation and immune cell recruitment, such as macrophages, further amplify this process. The persistent inflammation and oxidative stress promote DNA damage, genetic mutations, and genomic instability, ultimately leading to lung carcinogenesis.</p>
Full article ">Figure 6
<p>Role of NLRP3 inflammasome activation in promoting colon cancer development. During inflammatory bowel disease (IBD), such as Crohn’s disease and ulcerative colitis, the inflammation of the gut lining activates immune cells, causing oxidative stress, mitochondrial damage, and the release of DAMPs. Gut microbiome imbalance (dysbiosis) leads to pathogenic bacterial growth and loss of gut-barrier integrity, allowing pathogen and toxin leakage, further driving oxidative stress. These pathways cause NLRP3 activation and trigger IL-1β and IL-18 release. Increased inflammasome response results in immune system imbalance, epithelial-barrier dysfunction, and neoplasia initiation, ultimately contributing to colon cancer growth and metastasis.</p>
Full article ">
11 pages, 2211 KiB  
Article
A Novel Triplet of Alisertib Plus Ibrutinib Plus Rituximab Is Active in Mantle Cell Lymphoma
by Baskaran Subramani, Patrick J. Conway, Aisha Al-Khinji, Kun Zhang, Ritu Pandey and Daruka Mahadevan
Cancers 2024, 16(24), 4257; https://doi.org/10.3390/cancers16244257 (registering DOI) - 21 Dec 2024
Viewed by 252
Abstract
Background/Objectives: Aurora (AK) A/B are oncogenic mitotic kinases that when over-expressed are poor prognostic markers in mantle cell lymphoma (MCL). Methods and Results: Alisertib, an AK-A inhibitor, has anti-tumor activity in relapsed/refractory (r/r) MCL patients. We evaluated alisertib plus ibrutinib in [...] Read more.
Background/Objectives: Aurora (AK) A/B are oncogenic mitotic kinases that when over-expressed are poor prognostic markers in mantle cell lymphoma (MCL). Methods and Results: Alisertib, an AK-A inhibitor, has anti-tumor activity in relapsed/refractory (r/r) MCL patients. We evaluated alisertib plus ibrutinib in MCL to abrogate ibrutinib resistance. Alisertib plus ibrutinib was therapeutically synergistic on both Granta-519 insensitive to ibrutinib and JeKo-1 cells sensitive to ibrutinib. Alisertib decreased PI-3K, BTK, p38, HCK, and RSK kinases, indicative of its multipotent effect on cellular proliferation and growth. A mouse xenograft model of Granta-519 demonstrated that alisertib plus ibrutinib had a comparable anti-tumor response to ibrutinib plus rituximab. However, alisertib plus ibrutinib plus rituximab demonstrated significantly stronger tumor growth inhibition than the doublets. Conclusions: Both double and triple combinations showed enhanced survival versus ibrutinib alone. Ibrutinib insensitivity can be disrupted by alisertib plus ibrutinib in MCL. Full article
Show Figures

Figure 1

Figure 1
<p>MTS cell proliferation assay of MCL cell lines ((<b>A</b>–<b>C</b>) Granta-519; (<b>D</b>–<b>F</b>) JeKo-1). (<b>A</b>) IC50 of alisertib = 23.7 nM. (<b>B</b>) IC50 of Ibrutinib = 14,350 nM. (<b>C</b>) Leftward shift of concentration–effect curve for alisertib plus ibrutinib indicates significant synergism with an IC50 = 12.52 nM. (<b>D</b>) IC50 of alisertib = 3.01 nM. (<b>E</b>) IC50 of ibrutinib = 556.4 nM. (<b>F</b>) Ibrutinib and alisertib are synergistic in JeKo-1 inhibition.</p>
Full article ">Figure 2
<p>(<b>A</b>) Granta-519 cells and (<b>B</b>) Jeko-1 cells treated with alisertib in DMSO, 0, 1, 25, 50, 75, and 100 nM alisertib for 24 hrs. Western blotting of these cells showed decreased protein levels of BTK, PI3K, c-Myc, NF-kβ, and Bcl-2 in a dose-dependent manner. Quantification of signal intensity of the blots (<a href="#app1-cancers-16-04257" class="html-app">Supplementary Figure S2A</a> Granta-519 and <a href="#app1-cancers-16-04257" class="html-app">Figure S2B</a> Jeko-1, <a href="#app1-cancers-16-04257" class="html-app">Figure S4 in Supplementary data</a> have the uncropped Western blots).</p>
Full article ">Figure 3
<p>Phospho-protein array of Granta-519 cells treated with alisertib 1.0 μM for 7 days to optimize polyploidy showed no significant changes in p53 phosphorylation at Ser-15, Ser-46, or Ser-392. Of the kinases, decreased phosphorylation with alisertib was observed with HCK &gt; RSK 1/2/3 &gt; p38. Phospho-protein array is displayed as a percentage of the pixel density from 2 biologic replicates.</p>
Full article ">Figure 4
<p>Gene enrichment analysis of differentially expressed genes for pathways and molecular function pre-/post-alisertib treatment of Granta-519 (BCR-low) cells. (<b>A</b>) Altered signaling pathways by upregulated genes. (<b>B</b>) Gene ontology molecular functions for upregulated genes. (<b>C</b>) Downregulated cellular processes.</p>
Full article ">Figure 5
<p>Granta-519 xenograft SCID mouse model of MCL. (<b>A</b>) Tumor growth inhibition: N = 12 mice per cohort treated with ibrutinib, ibrutinib plus alisertib, ibrutinib plus rituximab, and triple therapy versus control. On day 14, the differences between the tumor burdens of the control group and each of the treatment groups were statistically significant (GraphPad Prism version 8.4.1). (<b>B</b>) Kaplan–Meier survival curves of control vs. ibrutinib vs. ibrutinib + alisertib vs. ibrutinib + rituximab and ibrutinib + alisertib + rituximab. There was no significant difference between control and ibrutinib-treated mice (<span class="html-italic">p</span> = 0.6258). There was a significant difference between the control and doublet- and triplet-treated mice (** <span class="html-italic">p</span> &lt; 0.0001) and between the ibrutinib- and combination-treated mice (* <span class="html-italic">p</span> &lt; 0.0001). There was no significant difference between the different combination treatments (<span class="html-italic">p</span> ≥ 0.589). <span class="html-italic">p</span>-values were determined by log-rank analysis.</p>
Full article ">
18 pages, 1019 KiB  
Article
Bacterial Outer Membrane Vesicle (OMV)-Encapsulated TiO2 Nanoparticles: A Dual-Action Strategy for Enhanced Radiotherapy and Immunomodulation in Oral Cancer Treatment
by Shun-An Kan, Li-Wen Zhang, Yu-Chi Wang, Cheng-Yu Chiang, Mei-Hsiu Chen, Shih-Hao Huang, Ming-Hong Chen and Tse-Ying Liu
Nanomaterials 2024, 14(24), 2045; https://doi.org/10.3390/nano14242045 (registering DOI) - 20 Dec 2024
Viewed by 215
Abstract
Oral squamous-cell carcinoma (OSCC) poses significant treatment challenges due to its high recurrence rates and the limitations of current therapies. Titanium dioxide (TiO2) nanoparticles are promising radiosensitizers, while bacterial outer membrane vesicles (OMVs) are known for their immunomodulatory properties. This study [...] Read more.
Oral squamous-cell carcinoma (OSCC) poses significant treatment challenges due to its high recurrence rates and the limitations of current therapies. Titanium dioxide (TiO2) nanoparticles are promising radiosensitizers, while bacterial outer membrane vesicles (OMVs) are known for their immunomodulatory properties. This study investigates the potential of OMV-encapsulated TiO2 nanoparticles (TiO2@OMV) to combine these effects for improved OSCC treatment. TiO2 nanoparticles were synthesized using a hydrothermal method and encapsulated within OMVs derived from Escherichia coli. The TiO2@OMV carriers were evaluated for their ability to enhance radiosensitivity and stimulate immune responses in OSCC cell lines. Reactive oxygen species (ROS) production, macrophage recruitment, and selective cytotoxicity toward cancer cells were assessed. TiO2@OMV demonstrated significant radiosensitization and immune activation compared to unencapsulated TiO2 nanoparticles. The system selectively induced cytotoxicity in OSCC cells, sparing normal cells, and enhanced ROS generation and macrophage-mediated antitumor responses. This study highlights TiO2@OMV as a dual-action therapeutic platform that synergizes radiotherapy and immunomodulation, offering a targeted and effective strategy for OSCC treatment. The approach could improve therapeutic outcomes and reduce the adverse effects associated with conventional therapies. Full article
(This article belongs to the Section Biology and Medicines)
21 pages, 4656 KiB  
Article
Synergistic Anti-Cancer Effects of Curcumin and Thymoquinone Against Melanoma
by Hana Mohd and Bozena Michniak-Kohn
Antioxidants 2024, 13(12), 1573; https://doi.org/10.3390/antiox13121573 - 20 Dec 2024
Viewed by 351
Abstract
Combining anti-cancer agents in cancer therapies is becoming increasingly common because of their improved efficacy, reduced toxicity, and decreased risk of resistance development. Melanoma, a highly aggressive form of skin cancer characterized by limited treatment options due to chemoresistance, poses a considerable challenge [...] Read more.
Combining anti-cancer agents in cancer therapies is becoming increasingly common because of their improved efficacy, reduced toxicity, and decreased risk of resistance development. Melanoma, a highly aggressive form of skin cancer characterized by limited treatment options due to chemoresistance, poses a considerable challenge for effective management. Here, we test the hypothesis that dietary supplements such as thymoquinone (TQ) and curcumin (CU) cooperatively modulate cancer-associated cellular mechanisms to inhibit melanoma progression. Through a series of in vitro experiments utilizing the A375 melanoma cell line, including assessments of cell viability, apoptosis, multicellular tumor spheroid models, reactive oxygen species (ROS) quantification, metabolomics analysis, and RNA sequencing, we established that the combined application of TQ and CU exhibited superior anti-tumor effects compared to their individual use. Our results indicate that the combination treatment significantly inhibited cell viability and induced apoptosis more effectively than either agent alone, with optimal synergy observed at concentrations of 25 µM CU and 10 µM TQ against A375 cells. Additionally, the combination treatment markedly elevated ROS levels, selectively activating the mitochondrial apoptotic pathway via caspase-9. Differential gene expression analysis further revealed a unique synergistic effect of the combination treatment, with enhanced regulation of genes related to oxidative stress and apoptosis. Notably, pathways such as mitochondrial apoptotic signaling and redox homeostasis were more effectively influenced by the combination, with genes such as GPX3, CYP4F11, and HSPB8 cooperatively regulated. Overall, the findings suggest that, in combination, TQ and CU acts synergistically against melanoma; however, further experimental and clinical studies are required to confirm its therapeutic potential. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of thymoquinone, curcumin, and their combination on the viability of human (A375) metastatic melanoma cells and the two healthy human keratinocyte (HaCaT) and human dermal fibroblast (HDfa) cell lines. The values are presented as the percentage of cell death. The data represent the mean ± SD of three independent experiments conducted in triplicate (vs. control, * <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 2
<p>Synergism between TQ and CU in A375 cells. Bliss and HSA synergy scores (SynergyFinder.com) were calculated to predict the potential synergism of TQ and CU.</p>
Full article ">Figure 3
<p>(<b>A</b>) Representative histograms obtained using flow cytometry on an apoptosis assay from various groups are shown (quadrant: upper left—dead cells; upper right—late apoptosis; lower left—live cells; lower right—early apoptosis); (<b>B</b>) percentage of viable, dead, early apoptotic, and late apoptotic cells in the control and compound-treated groups. <span class="html-italic">n</span> = 3; value are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control cells; ** <span class="html-italic">p</span> &lt; 0.05 combination vs. 25 μM CU; *** <span class="html-italic">p</span> &lt; 0.05 combination vs. 10 μM TQ.</p>
Full article ">Figure 4
<p>(<b>A</b>) Calcein-AM/PI dual-stained A375 multicellular tumor spheroids after 24 h of treatment with CU, TQ, or their combination. A red signal indicates dead cells, and a green signal indicates viable cells. Images obtained using an ECHO Revolve Microscope (Model RVL-100-M, Serial #: M-00395-RVL) (20×/0.4 objective). Scale bar: 180 µM. (<b>B</b>) Effect of thymoquinone, curcumin, and their combination on the viability of human (A375) metastatic melanoma cells in an MCTS model using a CellTiter-Glo 3D assay.<span class="html-italic">p</span>-Values are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control.</p>
Full article ">Figure 4 Cont.
<p>(<b>A</b>) Calcein-AM/PI dual-stained A375 multicellular tumor spheroids after 24 h of treatment with CU, TQ, or their combination. A red signal indicates dead cells, and a green signal indicates viable cells. Images obtained using an ECHO Revolve Microscope (Model RVL-100-M, Serial #: M-00395-RVL) (20×/0.4 objective). Scale bar: 180 µM. (<b>B</b>) Effect of thymoquinone, curcumin, and their combination on the viability of human (A375) metastatic melanoma cells in an MCTS model using a CellTiter-Glo 3D assay.<span class="html-italic">p</span>-Values are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control.</p>
Full article ">Figure 5
<p>(<b>A</b>) Intracellular ROS levels in A375 cells after treatment with CU, TQ, and their combination. After the indicated treatment for 24 h, cells were incubated with 10 μM DCFH-DA for 30 min and then immediately subjected to a flow cytometry analysis. The results are expressed as a ratio of the relative fluorescence intensity compared to the control group. (<b>B</b>) The DCFH-DA spectrum represents the fluorescence intensities of the probe. DCFH-DA (10 μM) was incubated with the cell culture for 30 min. (<b>C</b>) Caspases-9 and -8 were measured using Caspase-Glo 8 Assay and Caspase-Glo 9 Assay kits. <span class="html-italic">p</span>-Values were recorded as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control cells; ** <span class="html-italic">p</span> &lt; 0.05 combination vs. 25 μM CU; *** <span class="html-italic">p</span> &lt; 0.05 combination vs. 10 μM TQ.</p>
Full article ">Figure 5 Cont.
<p>(<b>A</b>) Intracellular ROS levels in A375 cells after treatment with CU, TQ, and their combination. After the indicated treatment for 24 h, cells were incubated with 10 μM DCFH-DA for 30 min and then immediately subjected to a flow cytometry analysis. The results are expressed as a ratio of the relative fluorescence intensity compared to the control group. (<b>B</b>) The DCFH-DA spectrum represents the fluorescence intensities of the probe. DCFH-DA (10 μM) was incubated with the cell culture for 30 min. (<b>C</b>) Caspases-9 and -8 were measured using Caspase-Glo 8 Assay and Caspase-Glo 9 Assay kits. <span class="html-italic">p</span>-Values were recorded as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control cells; ** <span class="html-italic">p</span> &lt; 0.05 combination vs. 25 μM CU; *** <span class="html-italic">p</span> &lt; 0.05 combination vs. 10 μM TQ.</p>
Full article ">Figure 6
<p>Significantly differentially expressed genes were clustered by their gene ontology, and the enrichment of gene ontology terms was tested using Fisher’s exact test (GeneSCF v1.1-p2): (<b>A</b>) significantly enriched gene ontology terms with adjusted <span class="html-italic">p</span>-values less than 0.05 in the differentially expressed gene sets (up to 40 terms) between 10 μM TQ+ 25 μM CU and the control; (<b>B</b>) mRNA expression of highly regulated genes, measured by RT-PCR and shown as the fold change compared to the control group. <span class="html-italic">p</span>-Values are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control.</p>
Full article ">Figure 7
<p>A bi-clustering heatmap for the combination vs. the control, which visualizes the expression profiles of the top 30 differentially expressed genes sorted by their adjusted <span class="html-italic">p</span>-values by plotting their log2-transformed expression values in the samples.</p>
Full article ">Figure 8
<p>Metabolomics of TQ, CU, and their combination on treated melanoma cell lines: (<b>A</b>) PCA analysis of three groups showing that samples in each group are clustered away from one another; (<b>B</b>) heatmap of pairwise correlation values of 120 metabolites and depiction of the major metabolic pathways in A375 cells; (<b>C</b>) identified pathways altered by the combination treatment vs. the control treatment in A375 cell lines.</p>
Full article ">Figure 8 Cont.
<p>Metabolomics of TQ, CU, and their combination on treated melanoma cell lines: (<b>A</b>) PCA analysis of three groups showing that samples in each group are clustered away from one another; (<b>B</b>) heatmap of pairwise correlation values of 120 metabolites and depiction of the major metabolic pathways in A375 cells; (<b>C</b>) identified pathways altered by the combination treatment vs. the control treatment in A375 cell lines.</p>
Full article ">Figure 8 Cont.
<p>Metabolomics of TQ, CU, and their combination on treated melanoma cell lines: (<b>A</b>) PCA analysis of three groups showing that samples in each group are clustered away from one another; (<b>B</b>) heatmap of pairwise correlation values of 120 metabolites and depiction of the major metabolic pathways in A375 cells; (<b>C</b>) identified pathways altered by the combination treatment vs. the control treatment in A375 cell lines.</p>
Full article ">
20 pages, 4965 KiB  
Review
Application of Metal Oxide Nanoparticles in Different Carcinomas
by Nutan Rani, Yousuf Khan, Sapna Yadav, Kalawati Saini and Dipak Maity
J. Nanotheranostics 2024, 5(4), 253-272; https://doi.org/10.3390/jnt5040015 - 20 Dec 2024
Viewed by 387
Abstract
Metal oxide nanoparticles (MONPs) have recently attracted much attention from researchers due to their use in cancer chemotherapy, targeted drug delivery, and diagnosis/MRI imaging. Various studies have demonstrated that different metal oxide NPs show cytotoxic effects by inducing apoptosis in cancerous cells and [...] Read more.
Metal oxide nanoparticles (MONPs) have recently attracted much attention from researchers due to their use in cancer chemotherapy, targeted drug delivery, and diagnosis/MRI imaging. Various studies have demonstrated that different metal oxide NPs show cytotoxic effects by inducing apoptosis in cancerous cells and do not have any toxic impact on normal cells. The mechanism of cytotoxicity is shown through reactive oxygen species (ROS) generated by (MONPs) in the cancerous cell. In vitro and in vivo studies reveal that in some cases metal oxide NPs are used alone and somewhere these NPs are used in combination with other therapies such as photodynamic therapy and with anticancer nanomedicines as drug carriers or drug conjugates. The phenomenon of enhanced permeability and retention (EPR) effect has been the basis of targeted drug delivery to cancerous tumors. Finally, we also provide a simple and comparative analysis of the major apoptosis pathways proposed to increase beginner understanding of anti-cancer nanomaterials. Herein, we have reviewed the most important antitumor results obtained with different metal oxide nanoparticles such as ZnO, Fe2O3/Fe3O4, CuO/Cu2O, TiO2, CeO2, and HfO2, respectively. These NPs can be applied to treat cancer by either passive or active processes. A passive process uses the enhanced permeability and retention (EPR) effect. Superparamagnetic iron oxide nanoparticles (SPIONs), due to their unique magnetic and physiochemical properties have been used in magnetic fluid hyperthermia (MFH) and magnetic resonance imaging (MRI) in vitro as well as in vivo. Now, the research has reached the stage of clinical trials for the treatment of various types of cancer. ZnO NPs have been used very vastly in cytotoxic as well as in targeted drug delivery. These NPs are also used for loading anticancer drugs such as doxorubicin. Herein, in this review, we have examined current advances in utilizing MONPs and their analogs as cancer therapeutic, diagnostic, and drug-delivery agents. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Six pathological hallmarks of cancer.</p>
Full article ">Figure 2
<p>Oncological applications of metal-oxide nanoparticles.</p>
Full article ">Figure 3
<p>Schematic representation of Lipo-IONP/DOX for combined chemo-photothermal cancer therapy [<a href="#B13-jnt-05-00015" class="html-bibr">13</a>] (open access).</p>
Full article ">Figure 4
<p>Represented the working principle of AuNCs@MnO<sub>2</sub> (AM) nanomaterial for ICD (immunogenic cell death) (<b>a</b>) synthesis procedure for AM and generation of O<sub>2</sub> (<b>b</b>) Therapeutic approach of AM for inducing ICD by stimulating more dead tumor cells (<b>c</b>–<b>e</b>) Identification of different ICD signal molecules during AM and laser treatments (<b>c</b>) fluorescence microscopy images showing CRT expression in 4T1 cells (<b>d</b>,<b>e</b>) liberated ATP and HMGB1 in the supernatant (<b>f</b>) diagrammatic illustration of ICD-induced DC activation (<b>g</b>) CD83 and CD86 expression after DC maturation (<b>h</b>) liberated IL-12 in the culture supernatant. Asterisks (*) denote statistically significant differences between PBS and other treatments. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. # <span class="html-italic">p</span> &lt; 0.05 (n = 5) indicates that the differences between the two groups are statistically significant. Reproduced with permission from [<a href="#B14-jnt-05-00015" class="html-bibr">14</a>], Copyright 2018, Elsevier.</p>
Full article ">Figure 5
<p>Major apoptosis mechanisms triggered by nanoparticles.</p>
Full article ">Figure 6
<p>SPIONs are administered intravenously (IV) for multimodal diagnostic and/or therapeutic uses [<a href="#B50-jnt-05-00015" class="html-bibr">50</a>] (open access).</p>
Full article ">
20 pages, 1801 KiB  
Article
Bioactive Molecules from the Exoskeleton of Procambarus clarkii: Reducing Capacity, Radical Scavenger, and Antitumor and Anti-Inflammatory Activities
by Francesco Longo, Francesca Di Gaudio, Alessandro Attanzio, Laura Marretta, Claudio Luparello, Serena Indelicato, David Bongiorno, Giampaolo Barone, Luisa Tesoriere, Ilenia Concetta Giardina, Giulia Abruscato, Manuela Perlotti, Lucie Branwen Hornsby, Vincenzo Arizza, Mirella Vazzana, Federico Marrone, Aiti Vizzini, Chiara Martino, Dario Savoca, Vinicius Queiroz, Antonio Fabbrizio and Manuela Mauroadd Show full author list remove Hide full author list
Biomolecules 2024, 14(12), 1635; https://doi.org/10.3390/biom14121635 - 20 Dec 2024
Viewed by 381
Abstract
This study evaluates, for the first time, the reducing capacity, radical scavenger activity, and in vitro antitumor and anti-inflammatory effects of chitosan, astaxanthin, and bio-phenols extracted from the exoskeleton of Sicilian Procambarus clarkii, the most widespread species of invasive crayfish in the [...] Read more.
This study evaluates, for the first time, the reducing capacity, radical scavenger activity, and in vitro antitumor and anti-inflammatory effects of chitosan, astaxanthin, and bio-phenols extracted from the exoskeleton of Sicilian Procambarus clarkii, the most widespread species of invasive crayfish in the Mediterranean region. Among the extracted compounds, astaxanthin exhibited the highest antioxidant activity in all assays. Chitosan and polyphenols demonstrated reducing and radical scavenging activity; chitosan showed significant ferric ion reducing capacity in the FRAP test, while bio-phenolic compounds displayed notable radical scavenging activity in the DPPH and ABTS assays. Both astaxanthin and polyphenols showed dose-dependent cytotoxicity on two different cancer cell lines, with IC50 values of 1.45 µg/mL (phenolic extract) and 4.28 µg/mL (astaxanthin extract) for HepG2 cells and 2.45 µg/mL (phenolic extract) and 4.57 µg/mL (astaxanthin extract) for CaCo-2 cells. The bio-phenolic extract also showed potential anti-inflammatory effects in vitro by inhibiting nitric oxide production in inflamed RAW 264.7 macrophages, reducing the treated/control NO ratio to 77% and 74% at concentrations of 1.25 and 1.5 μg/mL, respectively. These results suggest that P. clarkii exoskeletons could be a valuable source of bioactive molecules for biomedical, pharmaceutical, and nutraceutical application while contributing to the sustainable management of this invasive species. Full article
Show Figures

Figure 1

Figure 1
<p>IR spectrum of chitosan extracted from <span class="html-italic">P. clarkii</span>.</p>
Full article ">Figure 2
<p>HPLC/MS chromatogram and MS/MS spectrum of the peak 13.58 min from the ethanolic <span class="html-italic">P. clarkii</span> extract.</p>
Full article ">Figure 3
<p>Dose–response effect of phenolic extract from <span class="html-italic">P. clarkii</span> exoskeleton on the viability of HepG2 and CaCo-2 cells after 24 h of exposure. The error bars correspond to the standard error of the mean (s.e.m.) of three independent measurements. <span class="html-italic">p</span> values comparing viability ratios to controls were &lt;0.05 for every measurement.</p>
Full article ">Figure 4
<p>Dose–response effect of 24 h and 7 days of extraction of astaxanthin extract from <span class="html-italic">P. clarkii</span> exoskeleton on the viability of HepG2 (<b>A</b>) and CaCo-2 cells (<b>B</b>) after 24 h of exposure. The error bars correspond to the standard error of the mean (s.e.m.) of three independent measurements. <span class="html-italic">p</span> values comparing viability ratios to controls were &lt;0.05 for every measurement.</p>
Full article ">Figure 5
<p>Effect of astaxanthin (<b>A</b>) and polyphenol extract (<b>B</b>) from <span class="html-italic">P. clarkii</span> exoskeleton on the viability of RAW 264.7 macrophages after 24 h of exposure. The error bars correspond to the standard error of the mean (s.e.m.) of three independent measurements. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Effect of astaxanthin (<b>A</b>) and polyphenol extract (<b>B</b>) from <span class="html-italic">P. clarkii</span> exoskeleton on NO production by RAW 264.7 macrophages after 24 h of co-exposure with LPS compared with LPS alone. The error bars correspond to the standard error of the mean (s.e.m.) of three independent measurements. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
17 pages, 1347 KiB  
Article
A Novel Glycoengineered Humanized Antibody Targeting DLK1 Exhibits Potent Anti-Tumor Activity in DLK1-Expressing Liver Cancer Cell Xenograft Models
by Koji Nakamura, Kota Takahashi, Izumi Sakaguchi, Takumi Satoh, Lingyi Zhang, Hiroyuki Yanai and Yukihito Tsukumo
Int. J. Mol. Sci. 2024, 25(24), 13627; https://doi.org/10.3390/ijms252413627 - 19 Dec 2024
Viewed by 424
Abstract
Delta-like 1 homolog (DLK1), a non-canonical Notch ligand, is highly expressed in various malignant tumors, especially in hepatocellular carcinoma (HCC). CBA-1205 is an afucosylated humanized antibody against DLK1 with enhanced antibody-dependent cellular cytotoxicity (ADCC). The binding characteristics of CBA-1205 were analyzed by enzyme-linked [...] Read more.
Delta-like 1 homolog (DLK1), a non-canonical Notch ligand, is highly expressed in various malignant tumors, especially in hepatocellular carcinoma (HCC). CBA-1205 is an afucosylated humanized antibody against DLK1 with enhanced antibody-dependent cellular cytotoxicity (ADCC). The binding characteristics of CBA-1205 were analyzed by enzyme-linked immunosorbent assay and fluorescence-activated cell sorting assay. The ADCC activity of CBA-1205 was assessed. The anti-tumor efficacy of CBA-1205 was evaluated in xenograft mouse models, and toxicity and toxicokinetic profiles of CBA-1205 were evaluated in cynomolgus monkeys. CBA-1205 selectively bound to DLK1 among the Notch ligands and only to monkey and human DLK1. The binding epitope was between epidermal growth factor-like domains 1 and 2 of DLK1, which are not involved in any known physiological functions. The ADCC activity of CBA-1205 was confirmed using human peripheral blood mononuclear cells as effector cells. CBA-1205 as a single agent and in combination with lenvatinib demonstrated long-lasting anti-tumor efficacy, including tumor regression, in two liver cancer xenograft models. The toxicity and toxicokinetic profiles of CBA-1205 in cynomolgus monkeys were favorable. These findings suggest that CBA-1205 has the potential to be a useful therapeutic option for drug treatment in HCC. A phase 1 study is ongoing in patients with advanced cancers (jRCT2080225288, NCT06636435). Full article
(This article belongs to the Special Issue New Wave of Cancer Therapeutics: Challenges and Opportunities)
24 pages, 2081 KiB  
Article
Antitumor Activity and Multi-Target Mechanism of Phenolic Schiff Bases Bearing Methanesulfonamide Fragments: Cell Cycle Analysis and a Molecular Modeling Study
by Alaa A.-M. Abdel-Aziz, Adel S. El-Azab, Simone Brogi, Rezk R. Ayyad, Ibrahim A. Al-Suwaidan and Mohamed Hefnawy
Int. J. Mol. Sci. 2024, 25(24), 13621; https://doi.org/10.3390/ijms252413621 - 19 Dec 2024
Viewed by 248
Abstract
Five phenolic Schiff bases (711) incorporating a fragment of methanesulfonamide were synthesized and evaluated for their efficacy as antitumor agents. Compounds 7 and 8 demonstrated the most potent antitumor action, with a positive cytotoxic effect (PCE) of 54/59 and [...] Read more.
Five phenolic Schiff bases (711) incorporating a fragment of methanesulfonamide were synthesized and evaluated for their efficacy as antitumor agents. Compounds 7 and 8 demonstrated the most potent antitumor action, with a positive cytotoxic effect (PCE) of 54/59 and 59/59 and a mean growth percentage (MG%) of 67.3% and 19.5%, respectively, compared with imatinib (PCE = 20/59 and MG% = 92.6%). The PCE values for derivatives 911 were 3/59, 4/59, and 4/59, respectively, indicating poor antitumor effect. Compound 8 exhibited the most significant efficacy, suppressing cell proliferation by an average of 50% at a dosage of 0.501 µM, in comparison with the reference drugs sorafenib (2.33 µM), gefitinib (2.10 µM), erlotinib (7.68 µM), and celecoxib (17.5 µM). Compounds 7 and 8 had substantial inhibitory effects on the human epidermal growth factor receptor 2 (HER2), with IC50 values of 0.183 μM and 0.464 μM, respectively. Furthermore, they exhibited significant inhibition of the epidermal growth factor receptor (EGFR), with IC50 values of 0.752 μM and 0.166 μM, respectively. Compound 8 exhibited the highest COX-2 inhibition (IC50 = 12.76 μM). We performed molecular docking dynamic experiments to examine the precise interaction and structural prerequisites for the anticancer activity of derivatives 7 and 8 by targeting EGFR and HER2. Full article
(This article belongs to the Special Issue Design, Synthesis and Applications of the Task-Specific Molecules)
16 pages, 2709 KiB  
Article
PD1-Targeted Transgene Delivery to Treg Cells
by Vladislav A. Zhuchkov, Yulia E. Kravchenko, Elena I. Frolova and Stepan P. Chumakov
Viruses 2024, 16(12), 1940; https://doi.org/10.3390/v16121940 - 19 Dec 2024
Viewed by 378
Abstract
Achieving the precise targeting of lentiviral vectors (LVs) to specific cell populations is crucial for effective gene therapy, particularly in cancer treatment where the modulation of the tumor microenvironment can enhance anti-tumor immunity. Programmed cell death protein 1 (PD-1) is overexpressed on activated [...] Read more.
Achieving the precise targeting of lentiviral vectors (LVs) to specific cell populations is crucial for effective gene therapy, particularly in cancer treatment where the modulation of the tumor microenvironment can enhance anti-tumor immunity. Programmed cell death protein 1 (PD-1) is overexpressed on activated tumor-infiltrating T lymphocytes, including regulatory T cells that suppress immune responses via FOXP3 expression. We developed PD1-targeted LVs by incorporating the anti-PD1 nanobody nb102c3 into receptor-blinded measles virus H and VSV-Gmut glycoproteins. We assessed the retargeting potential of nb102c3 and evaluated transduction efficiency in activated T lymphocytes. FOXP3 expression was suppressed using shRNA delivered by these LVs. Our results demonstrate that PD1-targeted LVs exerted pronounced tropism towards PD1+ cells, enabling the selective transduction of activated T lymphocytes while sparing naive T cells. The suppression of FOXP3 in Tregs reduced their suppressive activity. PD1-targeted glycoprotein H provided greater specificity, whereas the VSV-Gmut, together with the anti-PD1 pseudoreceptor, achieved higher viral titers but was less selective. Our study demonstrates that PD1-targeted LVs may offer a novel strategy to modulate immune responses within the tumor microenvironment with the potential for developing new therapeutic strategies aimed at enhancing anti-tumor immunity. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic representations of retargeted measles (Hc∆18AA-102c3, 4AHc∆24AA-102c3), Nipah (Nip∆34Gm4-102c3), and VSV (VSVG<sub>mut</sub>+102c3R) glycoproteins tested for lentivector retargeting. (<b>B</b>) Predicted structure of the homodimer of the measles H-protein head fused with nb102c3. (<b>C</b>) Microphotographs of the syncytia formation test performed on HEK-293T and HEK-293T-PD1 cells transfected with Hc∆18AA-102c3, 4AHc∆24AA-102c3, and Nip∆34Gm4-102c3 with corresponding F-protein-encoding plasmids and the lentivector plasmid encoding tagRFP (red channel). (<b>D</b>) Infectious titers of all retargeted LVs (i.u.) and selectivity (fold). (<b>E</b>) FACS plots of HEK-293T and HEK-293T-PD1 cells transduced with a corresponding lentivector carrying the tagRFP sequence (1 mL of non-concentrated lentivirus sample per 35 mm well).</p>
Full article ">Figure 2
<p>(<b>A</b>) qPCR quantitation of PDCD1 levels in PBMCs cultured with IL-2 (ctrl), IL-2 + PHA-M (1), PMA + ionomycin + PHA-M (2), IL-2 + PHA-M + PMA + ionomycin (3), and IL-2 + PHA-M + PMA + ionomycin + IFNγ + IL-4 + IL-12 (4), **—<span class="html-italic">p</span> = 0.007. (<b>B</b>) nanoLuc luminescence levels normalized to cell count after transduction with lentiviral vectors of CD4<sup>+</sup> T cells with or without transduction enhancers (Treatment 3, as in (<b>A</b>), and Treatment 3-PB-F108), and in the presence of 50 µg/mL azidothymidine (Treatment 3 + AZT). (<b>C</b>) nanoLuc luminescence levels normalized to cell count after transduction with control (4AHc∆24) and targeted (4AHc∆24AA-102c3, VSVGmut+102c3R) lentivectors, **—<span class="html-italic">p</span> &lt; 0.0075. (<b>D</b>) nanoLuc luminescence levels in transduced CD4<sup>+</sup> T cells, **—<span class="html-italic">p</span> = 0.0018, ***—<span class="html-italic">p</span> = 0.0005.</p>
Full article ">Figure 3
<p>shRNA knockdown of FOXP3 with a non-targeted lentivector. (<b>A</b>) Schematic representation of shRNA-expressing lentivector and its cassette integrated in the genome. (<b>B</b>) qPCR quantitation of FOXP3 expression in stimulated CD4<sup>+</sup> T cells and Tregs transduced with shFOXP3 and control shRNA, **—<span class="html-italic">p</span> = 0.0065. (<b>C</b>) qPCR quantitation of FOXP3 expression in PD1<sup>+</sup> CD4<sup>+</sup> T cells and PD1<sup>+</sup> Tregs transduced with shFOXP3 and control shRNA, *—<span class="html-italic">p</span> = 0.015. (<b>D</b>) TGFβ levels in media of PD1<sup>−</sup> and PD1<sup>+</sup> Tregs transduced with shFOXP3 and the control, ****—<span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Cytometry measurements of CD4<sup>+</sup> PD1<sup>+</sup> T lymphocytes co-cultured with U937-tagGFP and transduced with 4AHc∆24AA-102c3-pseudotyped (<b>A</b>) VSVG<sub>mut</sub>+102c3R-pseudotyped (<b>B</b>) and 4AHc∆24-pseudotyped (<b>C</b>) LVs carrying the tagRFP sequence. (<b>D</b>) FOXP3 expression in PD1<sup>+</sup> Tregs co-cultured with U937 cells and transduced with different pseudotypes of LVs carrying shFOXP3 or control shRNA. *—<span class="html-italic">p</span> = 0.0485, ns—<span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">
15 pages, 7093 KiB  
Article
Synthesis of Novel Benzofuran Spiro-2-Pyrrolidine Derivatives via [3+2] Azomethine Ylide Cycloadditions and Their Antitumor Activity
by Bowen Pan, Tao Wang, Liangliang Zheng, Zhangchao Dong, Lijuan Liu, Xiongwei Liu, Tingting Feng, Ying Zhou and Yang Shi
Int. J. Mol. Sci. 2024, 25(24), 13580; https://doi.org/10.3390/ijms252413580 - 19 Dec 2024
Viewed by 232
Abstract
A synthetic strategy of a three-component spiro-pyrrolidine compound based on benzofuran via an [3+2] azomethine ylide cycloaddition reaction is reported herein. Under mild optimal conditions, this reaction can quickly produce potentially bioactive compounds with a wide range of substrates, high yield, and simple [...] Read more.
A synthetic strategy of a three-component spiro-pyrrolidine compound based on benzofuran via an [3+2] azomethine ylide cycloaddition reaction is reported herein. Under mild optimal conditions, this reaction can quickly produce potentially bioactive compounds with a wide range of substrates, high yield, and simple operation. The desired products were obtained with a yield of 74–99% and a diastereomeric ratio (dr) of >20:1. Subsequently, the inhibitory effects of the compounds on the cell viability of the human cancer cell line HeLa and mouse cancer cell line CT26 were evaluated. Compounds 4b (IC50 = 15.14 ± 1.33 µM) and 4c (IC50 = 10.26 ± 0.87 µM) showed higher antiproliferative activities against HeLa cells than cisplatin (IC50 = 15.91 ± 1.09 µM); compounds 4e (IC50 = 8.31 ± 0.64 µM) and 4s (IC50 = 5.28 ± 0.72 µM) exhibited better inhibitory activities against CT26 cells than cisplatin (IC50 = 10.27 ± 0.71 µM). The introduction of electron-donating substituents was beneficial to the inhibitory activities against cancer cells. Molecular docking simulations revealed that 4e and 4s may exert corresponding bioactivities by binding to antitumor targets through hydrogen bonds, providing a new approach for discovering spiro-heterocyclic antitumor drugs. Full article
(This article belongs to the Special Issue Recent Progress in Addition Reactions and Organic Synthesis)
Show Figures

Figure 1

Figure 1
<p>Benzofuran ring and spiro-pyrrolidine compounds [<a href="#B20-ijms-25-13580" class="html-bibr">20</a>,<a href="#B21-ijms-25-13580" class="html-bibr">21</a>,<a href="#B23-ijms-25-13580" class="html-bibr">23</a>].</p>
Full article ">Figure 2
<p>Structure of compound <b>4s</b> determined by X-ray crystallography.</p>
Full article ">Figure 3
<p>Mode of action of the protein TNFα with <b>4b</b> (<b>a</b>) and <b>4c</b> (<b>b</b>).</p>
Full article ">Figure 4
<p>Mode of action of the protein FTase with <b>4e</b> (<b>a</b>) and <b>4s</b> (<b>b</b>).</p>
Full article ">Scheme 1
<p>Substrate scope <sup>a</sup>. <sup>a</sup> The reaction was conducted using <b>1</b> (0.3 mmol), <b>2</b> (0.6 mmol), and <b>3</b> (0.9 mmol) in THF (3.0 mL) at 60 °C.; <sup>b</sup> Yield of isolated product is given. <sup>c</sup> Determined based on the yield ratio of isomers separated by column chromatography.</p>
Full article ">
18 pages, 2395 KiB  
Article
The Anticancer Activity of Cannabinol (CBN) and Cannabigerol (CBG) on Acute Myeloid Leukemia Cells
by Ahmad Kadriya, Sarah Forbes-Robertson and Mizied Falah
Molecules 2024, 29(24), 5970; https://doi.org/10.3390/molecules29245970 - 18 Dec 2024
Viewed by 284
Abstract
Several cannabis plant-derived compounds, especially cannabinoids, exhibit therapeutic potential in numerous diseases and conditions. In particular, THC and CBD impart palliative, antiemetic, as well as anticancer effects. The antitumor effects include inhibition of cancerous cell growth and metastasis and induction of cell death, [...] Read more.
Several cannabis plant-derived compounds, especially cannabinoids, exhibit therapeutic potential in numerous diseases and conditions. In particular, THC and CBD impart palliative, antiemetic, as well as anticancer effects. The antitumor effects include inhibition of cancerous cell growth and metastasis and induction of cell death, all mediated by cannabinoid interaction with the endocannabinoid system (ECS). However, the exact molecular mechanisms are still poorly understood. In addition, their effects on leukemia have scarcely been investigated. The current work aimed to assess the antileukemic effects of CBN and CBG on an acute monocytic leukemia cell line, the THP-1. THP-1 cell viability, morphology and cell cycle analyses were performed to determine potential cytotoxic, antiproliferative, and apoptotic effects of CBN and CBG. Western blotting was carried out to measure the expression of the proapoptotic p53. Both CBN and CBG inhibited cell growth and induced THP-1 cell apoptosis and cell cycle arrest in a dose- and time-dependent manner. CBN and CBG illustrated different dosage effects on THP-1 cells in the MTT assay (CBN > 40 μΜ, CBG > 1 μM) and flow cytometry (CBN > 5 μM, CBG > 40 μM), highlighting the cannabinoids’ antileukemic activity. Our study hints at a direct correlation between p53 expression and CBG or CBN doses exceeding 50 μM, suggesting potential activation of p53-associated signaling pathways underlying these effects. Taken together, CBG and CBN exhibited suppressive, cell death-inducing effects on leukemia cells. However, further in-depth research will be needed to explore the molecular mechanisms driving the anticancer effects of CBN and CBG in the leukemia setting. Full article
(This article belongs to the Topic Research on Natural Products of Medical Plants)
Show Figures

Figure 1

Figure 1
<p>The effect of CBN and CBG on THP-1 cell viability. (<b>A</b>,<b>C</b>) indicate the effect of various concentrations of CBN and CBG, respectively, on THP-1 cells after 24-h incubation. (<b>B</b>,<b>D</b>) indicate the effect of the increasing concentrations of CBN and CBG, respectively, on THP-1 cells after 48-h incubation. The star (*) indicates a <span class="html-italic">p</span>-value of <span class="html-italic">p</span> &lt; 0.05, which was considered statistically significant between the control (culture medium without treatment) and the increasing concentrations using CBG or CBN.</p>
Full article ">Figure 2
<p>The effects of CBN and CBG on the cell cycle of THP-1 cells. Chi-squared analysis was performed to assess the significance of differences between the control and the increasing concentrations of (<b>A</b>) CBN- or (<b>B</b>) CBG-treated cells. The star (*) represents a <span class="html-italic">p</span>-value &lt; 0.05 compared to the control (cell with culture medium only). The data shown here is a typical experiment repeated three times (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>The effects of CBN and CBG on the cell percentage of THP-1 cell population in SSC and FSC scatter analysis. The presented data are the average of triplicate samples in three different experiments.</p>
Full article ">Figure 4
<p>Effect of CBG or CBN treatment on THP-1 cell morphological characteristics. THP-1 cells were treated with CBN (<b>upper panel</b>) or CBG (<b>lower panel</b>) for 24 h at concentrations of 0 µM (<b>A</b>), 1 µM (<b>B</b>), 50 µM (<b>C</b>), and 100 µM (<b>D</b>). The red arrows mark changes in morphological characteristics in THP-1 cells in comparison with the control 0 µM (<b>A</b>). Representative fields were imaged at 200× magnification. Scale bar = 25 µm.</p>
Full article ">Figure 5
<p>Effect of CBG or CBN treatment on THP-1 cell morphological characteristics. THP-1 cells were treated with CBN (<b>upper panel</b>) or CBG (<b>lower panel</b>) for 48 h at concentrations of 0 µM (<b>A</b>), 1 µM (<b>B</b>), 50 µM (<b>C</b>), and 100 µM (<b>D</b>). The red arrows mark changes in morphological characteristics in THP-1 cells in comparison with the control 0 µM (<b>A</b>). Representative fields were imaged at 200× magnification. Scale bar = 25 µm.</p>
Full article ">Figure 6
<p>Western blot analysis of p53 expression in THP-1 leukemia cells following cannabinoid treatment. Expression of p53 in cells treated with CBN (<b>upper panel</b>) and CBG (<b>lower panel</b>) for 24 h with 0, 1, 50, and 100 μM is shown. Beta-actin bands were used as a loading control, as indicated in (<b>A</b>). Relative to the untreated control (0 μM), both CBG and CBN treatments induced an increase in p53 protein levels at 1, 50, and 100 μM, with maximal intensity observed at 50 and 100 μM. Notably, beta-actin expression was absent at 50 and 100 μM. The band’s volume intensity was quantified via the optical density of each band, as indicated in (<b>B</b>).</p>
Full article ">
21 pages, 5449 KiB  
Article
Rational Design of an Epidermal Growth Factor Receptor Vaccine: Immunogenicity and Antitumor Research
by Yifei Liu, Zehui Liu and Zhongliang Zheng
Biomolecules 2024, 14(12), 1620; https://doi.org/10.3390/biom14121620 - 18 Dec 2024
Viewed by 439
Abstract
The epidermal growth factor receptor (EGFR) is frequently overexpressed in a variety of human epithelial tumors, and its aberrant activation plays a pivotal role in promoting tumor growth, invasion, and metastasis. The clinically approved passive EGFR-related therapies have numerous limitations. Seven EGFR-ECD epitope [...] Read more.
The epidermal growth factor receptor (EGFR) is frequently overexpressed in a variety of human epithelial tumors, and its aberrant activation plays a pivotal role in promoting tumor growth, invasion, and metastasis. The clinically approved passive EGFR-related therapies have numerous limitations. Seven EGFR-ECD epitope peptides (EG1-7) were selected through bioinformatics epitope prediction tools including NetMHCpan-4.1, NetMHCIIpan-3.2, and IEDB Consensus (v2.18 and v2.22) and fused to the translocation domain of diphtheria toxin (DTT). The A549 tumor model was successfully established in a murine mouse model. The vaccine was formulated by combining the adjuvants Alum and CpG and subsequently assessed for its immunogenicity and anti-tumor efficacy. DTT-EG (3;5;6;7) vaccines elicited specific humoral and cellular immune responses and effectively suppressed tumor growth in both prophylactic and therapeutic mouse tumor models. The selected epitopes EG3 (HGAVRFSNNPALCNV145-159), EG5 (KDSLSINATNIKHFK346-360), EG6 (VKEITGFLLIQAWPE398-412), and EG7 (LCYANTINWKKLFGT469-483) were incorporated into vaccines for active immunization, representing a promising strategy for the treatment of tumors with overexpressed epidermal growth factor receptor (EGFR). The vaccine design and fusion method employed in this study demonstrate a viable approach toward the development of cancer vaccines. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Immunization with DTT-EG vaccines in combination with tumor cell injection protocol. (<b>A</b>) Prophylactic tumor model: tumor cell injection and vaccine immunization protocol (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a>) and (<b>B</b>) therapeutic tumor model: tumor cell injection and vaccine immunization protocol (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a> (accessed on 9 June 2024)).</p>
Full article ">Figure 2
<p>The predicted epitope EG in EGFR-ECD is demonstrated, along with the design and expression purification of DTT-EG utilizing DTT as a vector. (<b>A</b>) Displaying the EG epitope within EGFR-ECD (EGFR PDB id:3njp): EG1 is represented by a yellow sequence, EG2 by a red sequence, EG3 by a green sequence, EG4 by a blue and orange sequence, EG5 by an orange and purple sequence, EG6 by a cyan sequence, and EG7 by a pink sequence. (<b>B</b>) Design of DTT-EG tandem recombinant protein. DTT (202–373) denotes the amino acid fragment spanning from 202 to 373 of the DTT protein. The epitope prediction tool identified seven human-specific epitope peptides in the form of EG1, EG2, EG3, EG4, and E5G5G6G7 consisting of 15 amino acid residues each. GS represents the GS-linker sequence (GGTGGTGGTGGTAGTGGTGGTGGTGGTAGT). (<b>C</b>) Analysis of purified recombinant protein using 12% SDS-PAGE (M: Marker; A: DTT-EG1; B: DTT-EG2; C: DTT-EG3; D: DTT-EG4; E: DTT-EG5; F: DTT-EG6; G: DTT-EG7; H: DTT).</p>
Full article ">Figure 3
<p>Serum antibodies were detected using enzyme-linked immunosorbent assay (ELISA) following immunization of mice with the DTT-EG tandem recombinant protein. (Significance levels were denoted as ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, while “ns” indicated no significant difference). (<b>A</b>) Schematic representation of the immunization protocol for mice (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a> (accessed on 9 June 2024)). (<b>B</b>) ELISA analysis was conducted on the serum of immunized mice, with the coated proteins being DTT or EGFR. (<b>C</b>) The titer of antibodies against EGFR in the mouse serum following vaccination was determined by ELISA.</p>
Full article ">Figure 4
<p>The splenic lymphocytes of immunized mice were tested for cell proliferation, toxicity, and interferon-gamma release, and the spleens were tested for CD4 and CD8 immunohistochemistry. (Significance levels were denoted as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, while “ns” indicated no significant difference). (<b>A</b>) Schematic representation of the immunization protocol for mice (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a> (accessed on 9 June 2024)). (<b>B</b>) Cell proliferation detection experiment by CCK-8 method. (<b>C</b>) Cell cytotoxicity detection experiment by lactate dehydrogenase method. (<b>D</b>) Detection of IFN-γ release by ELISA method. (<b>E</b>) The spleen of immunized mice was stained by immunohistochemistry with anti-CD4 specific antibody. (<b>F</b>) The spleen of immunized mice was stained by immunohistochemistry with anti-CD8 specific antibody. (<b>G</b>) CD4+ T and CD8+ T-cell density was quantified using ImageJ.</p>
Full article ">Figure 5
<p>Antitumor effects of the DTT-EG vaccine in a prophylactic and therapeutic mouse A549 tumor model. (Significance levels were denoted as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, while “ns” indicated no significant difference). (<b>A</b>) prophylactic tumor model tumor growth curve, (<b>B</b>) prophylactic tumor model tumor weight, (<b>C</b>) therapeutic tumor model tumor growth curve, and (<b>D</b>) therapeutic tumor model tumor weight.</p>
Full article ">Figure 6
<p>The DTT-EG vaccine modulates the infiltration of CD8+ T cells and induces necrosis within the intratumoral tissue. (Significance levels were denoted as ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001, while “ns” indicated no significant difference). (<b>A</b>) Tumor tissues underwent immunohistochemical staining using an anti-CD4 specific antibody. (<b>B</b>) Tumor tissues underwent immunohistochemical staining using an anti-CD8 specific antibody. (<b>C</b>) CD4+ T and CD8+ T cell density was quantified using ImageJ. (<b>D</b>) Revealing the histopathological features of tumor tissue through hematoxylin and eosin (H&amp;E) staining.</p>
Full article ">
Back to TopTop