Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (72)

Search Parameters:
Keywords = tumoricidal activity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 3372 KB  
Article
Analysis of Immune Checkpoints on Peripheral Blood Mononuclear Cells Can Predict Clinical Outcome and Reveal Potential of HVEM-BTLA Axis in Epithelial Ovarian Cancers
by Yen-Ling Lai, Han-Wei Lin, Yu-Wen Huang, Jung Chen, Ming-Chien Tai, Chia-Ying Wu, Tyan-Shin Yang, Valentina Oblin, Kristin Shea and Yu-Li Chen
Pharmaceuticals 2025, 18(9), 1295; https://doi.org/10.3390/ph18091295 - 29 Aug 2025
Cited by 2 | Viewed by 1133
Abstract
Background/Objectives: Immune checkpoint inhibitors (ICIs) do not provide promising benefits to patients with advanced epithelial ovarian cancer (EOC). This study analyzed preoperative peripheral blood mononuclear cells (PBMCs) from these patients to evaluate the prognostic and therapeutic checkpoints. Methods: Preoperative PBMCs of 69 advanced [...] Read more.
Background/Objectives: Immune checkpoint inhibitors (ICIs) do not provide promising benefits to patients with advanced epithelial ovarian cancer (EOC). This study analyzed preoperative peripheral blood mononuclear cells (PBMCs) from these patients to evaluate the prognostic and therapeutic checkpoints. Methods: Preoperative PBMCs of 69 advanced EOC cases were collected to analyze the correlation between IC-expressing immune cells and survivals of patients. Co-expression of various ICs on the T lymphocytes from these patients was examined. Activation potential of programmed cell death 1 (PD-1)+herpes virus entry mediator (HVEM)+ T cells in PBMCs from the healthy donors and tumoricidal abilities of PMBCs treated with various ICIs were evaluated in vitro. Impact of respective ICIs on activation of T cells in PMBCs was investigated. Results: Percentages of PD-1+ CD4+ and CD8+ T cells in the PBMCs of patients could positively correlate with disease-free or overall survival. HVEM was highly co-expressed on these T lymphocytes. Prediction potential for overall survival of patients by the subpopulation of PD-1+ CD4+ or CD8+ T cells was higher than that by other parameters. The PD-1+HVEM+ CD4+ and CD8+ T cells showed characteristics of activated phenotype under activation signals. PBMCs receiving anti-B and T lymphocyte attenuator (BTLA) plus anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) or anti-PD-1 Ab had potent tumor-killing ability. Anti-BTLA Ab can drive T cells in the PBMCs toward an effector status. Conclusions: Percentages of PD-1+ T cells in the PBMCs could predict survival of EOC patients. Targeting HVEM-BTLA axis may be considered for ICI treatment of EOCs. Full article
(This article belongs to the Special Issue Tumor Immunopharmacology)
Show Figures

Figure 1

23 pages, 4480 KB  
Review
The Biophysics of Flash Radiotherapy: Tools for Measuring Tumor and Normal Tissues Microenvironment
by Islam G. Ali and Issam El Naqa
Antioxidants 2025, 14(8), 899; https://doi.org/10.3390/antiox14080899 - 23 Jul 2025
Cited by 1 | Viewed by 2305
Abstract
Ultra-high dose rate radiotherapy known as Flash radiotherapy (FLASH-RT) offers tremendous opportunities to improve the therapeutic ratio of radiotherapy by sparing the normal tissue while maintaining similar tumoricidal efficacy. However, the underlying biophysical basis of the FLASH effect remains under active investigation with [...] Read more.
Ultra-high dose rate radiotherapy known as Flash radiotherapy (FLASH-RT) offers tremendous opportunities to improve the therapeutic ratio of radiotherapy by sparing the normal tissue while maintaining similar tumoricidal efficacy. However, the underlying biophysical basis of the FLASH effect remains under active investigation with several proposed mechanisms involving oxygen depletion, altered free-radical chemistry, and differential biological responses. This article provides an overview of available experimental and computational tools that can be utilized to probe the tumor and normal tissue microenvironment. We analyze in vitro, ex vivo, and in vivo systems used to study FLASH responses. We describe various computational and imaging technologies that can potentially aid in understanding the biophysics of FLASH-RT and lead to safer clinical translational. Full article
(This article belongs to the Special Issue Oxidative Stress, Antioxidants, and Mechanisms in FLASH Radiotherapy)
Show Figures

Figure 1

16 pages, 2268 KB  
Article
Hydnocarpin, a Natural Flavonolignan, Induces the ROS-Mediated Apoptosis of Ovarian Cancer Cells and Reprograms Tumor-Associated Immune Cells
by Jae-Yoon Kim, Yejin Kim, Soo-Yeon Woo, Jin-Ok Kim, Hyunsoo Kim, So-Ri Son, Dae Sik Jang and Jung-Hye Choi
Antioxidants 2025, 14(7), 846; https://doi.org/10.3390/antiox14070846 - 10 Jul 2025
Cited by 1 | Viewed by 1251
Abstract
Ovarian cancer, the most lethal form of gynecological cancer worldwide with a poor prognosis, is largely driven by an immunosuppressive tumor microenvironment. In this study, we investigated the anticancer effects of hydnocarpin, a natural flavonolignan derived from the flowers of Pueraria lobata, [...] Read more.
Ovarian cancer, the most lethal form of gynecological cancer worldwide with a poor prognosis, is largely driven by an immunosuppressive tumor microenvironment. In this study, we investigated the anticancer effects of hydnocarpin, a natural flavonolignan derived from the flowers of Pueraria lobata, focusing on its effects on ovarian cancer and tumor-associated immune cells, including ovarian cancer-stimulated macrophages (MQs) and T cells. Hydnocarpin exhibited potent cytotoxicity against multiple ovarian cancer cell lines but only minimal toxicity against normal ovarian surface epithelial cells. Mechanistically, hydnocarpin triggered caspase-dependent apoptosis, as evidenced by the activation of caspase-9 and -3, with limited involvement of caspase-8, indicating the activation of the intrinsic apoptotic pathway. Experimental data implicated reactive oxygen species generation as a key mediator of hydnocarpin cytotoxicity, and reactive oxygen species inhibition significantly inhibited this cytotoxicity. In addition to its direct tumoricidal effects, hydnocarpin reprogrammed the tumor-associated immune cells, ovarian cancer-stimulated macrophages and T cells, by downregulating the levels of M2 MQ markers and pro-tumoral factors (matrix metalloproteinase-2/9, C–C motif chemokine ligand 5, transforming growth factor-β, and vascular endothelial growth factor) and enhancing MQ phagocytosis. Additionally, hydnocarpin promoted T-cell activation (interferon-γ and interleukin-2) and reduced the expression levels of immune evasion markers (CD80, CD86, and VISTA). Overall, this study demonstrated the dual anti-tumor effects of hydnocarpin on both ovarian cancer cells and immunosuppressive immune components in the tumor microenvironment, highlighting its potential as a novel therapeutic candidate for ovarian cancer. Full article
Show Figures

Graphical abstract

15 pages, 4481 KB  
Article
Nodal Expansion, Tumor Infiltration and Exhaustion of Neoepitope-Specific Th Cells After Prophylactic Peptide Vaccination and Anti-CTLA4 Therapy in Mouse Melanoma B16
by Alexandra V. Shabalkina, Anna V. Izosimova, Ekaterina O. Ryzhichenko, Elizaveta V. Shurganova, Daria S. Myalik, Sofia V. Maryanchik, Valeria K. Ruppel, Dmitriy I. Knyazev, Nadezhda R. Khilal, Ekaterina V. Barsova, Irina A. Shagina and George V. Sharonov
Int. J. Mol. Sci. 2025, 26(13), 6453; https://doi.org/10.3390/ijms26136453 - 4 Jul 2025
Cited by 1 | Viewed by 1069
Abstract
Peptide vaccines possess several advantages over mRNA vaccines but are generally less effective at inducing antitumor immunity. The bottlenecks limiting peptide vaccine efficacy could be elucidated by tracking and comparing vaccine-induced T-lymphocytes in successful and unsuccessful cases. Here we have applied our recent [...] Read more.
Peptide vaccines possess several advantages over mRNA vaccines but are generally less effective at inducing antitumor immunity. The bottlenecks limiting peptide vaccine efficacy could be elucidated by tracking and comparing vaccine-induced T-lymphocytes in successful and unsuccessful cases. Here we have applied our recent database of neoantigen-specific T cell receptors (TCRs) to profile tumor-specific T cells following vaccination with a neoantigen peptide vaccine and to correlate this with the response. Mice were vaccinated prophylactically with p30 peptide encoding B16 melanoma neoantigen (K739N mutation in Kif18b gene). The B16F0 melanoma in the vaccinated mice was additionally treated by a CTLA-4 checkpoint blockade. T cells from the tumors, tumor-draining lymph nodes (tdLNs) and vaccine depots were isolated, phenotyped, sorted by subsets and sequenced for TCR repertoires. The vaccine induced the accumulation of tumor-specific CD4+ Th cells in the tdLNs, while in the tumors these cells were present and their frequencies were not changed by the vaccine. These cells also accumulated at the vaccine depots, where they were phenotypically skewed by the vaccine components; however, these effects were minor due to approximately 50-fold lower cell quantities compared to the tdLNs. Only some of the p30-specific Th cells showed tumoricidal activity, as revealed by the reverse correlation of their frequencies in the tdLNs with the tumor size. The CTLA-4 blockade did not affect the tumor growth or the frequencies of tumor-specific cells but did stimulate Th cell motility. Thus, we have shown that tumor-specific Th clones accumulate and/or expand in the tdLNs, which correlates with tumor suppression but only for some of these clones. Tumor infiltration by these clones is not correlated with the growth rate. Full article
(This article belongs to the Special Issue New Insights in Tumor Immunity)
Show Figures

Figure 1

21 pages, 5329 KB  
Article
Development of Immune-Regulatory Pseudo-Protein-Coated Iron Oxide Nanoparticles for Enhanced Treatment of Triple-Negative Breast Tumor
by Ying Ji, Juan Li, Li Ma, Zhijie Wang, Bochu Du, Hiu Yee Kwan, Zhaoxiang Bian and Chih-Chang Chu
Nanomaterials 2025, 15(13), 1006; https://doi.org/10.3390/nano15131006 - 30 Jun 2025
Viewed by 1137
Abstract
Triple-negative breast cancer (TNBC) frequently evades immune recognition and elimination, resulting in an immunosuppressive microenvironment. The phagocytic activity of tumor-associated macrophages underscores the development of nanomaterials as a promising strategy to target these macrophages and modulate their polarization, thereby advancing immunotherapy against TNBC. [...] Read more.
Triple-negative breast cancer (TNBC) frequently evades immune recognition and elimination, resulting in an immunosuppressive microenvironment. The phagocytic activity of tumor-associated macrophages underscores the development of nanomaterials as a promising strategy to target these macrophages and modulate their polarization, thereby advancing immunotherapy against TNBC. This research developed functional polymers that are complexed with therapeutic molecules as a coating strategy for iron oxide nanoparticles. An arginine-based poly (ester urea urethane) polymer complexed with a macrophage-polarizing molecule (APU-R848) could provide a synergistic effect with iron oxide nanoparticles (IONPs) to stimulate the M1-polarization of macrophages at the tumor site, resulting in a versatile nano-platform for immune regulation of TNBC. In the 4T1 in vivo breast tumor model, the APU-R848-IONPs demonstrated an improved intratumoral biodistribution compared to IONPs without a polymer coating. APU-R848-IONPs significantly reversed the immune-suppressive tumor environment by reducing the M2/M1 macrophage phenotype ratio by 51%, associated with an elevated population of cytotoxic T cells and a significantly enhanced production of tumoricidal cytokines. The activated immune response induced by APU-R848-IONP resulted in a significant anti-tumor effect, demonstrating an efficacy that was more than 3.2-fold more efficient compared to the controls. These immune-regulatory pseudo-protein-coated iron oxide nanoparticles represent an effective nano-strategy for macrophages’ regulation and the activation of anti-tumor immunity, providing a new treatment modality for triple-negative breast cancer. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Figure 1

15 pages, 1915 KB  
Communication
Performance of Imidazoquinoline Glycoconjugate BAIT628 as a TLR7 Agonist Prodrug for Prostate Cancer
by Seyedeh A. Najibi, S. M. Al Muied Pranto, Muhammad Haroon, Amy E. Nielsen and Rock J. Mancini
Pharmaceuticals 2025, 18(6), 804; https://doi.org/10.3390/ph18060804 - 27 May 2025
Cited by 1 | Viewed by 2032
Abstract
Despite broad anti-cancer efficacy as Toll-Like Receptor (TLR) 7/8 agonists, imidazoquinolines remain limited in use via systemic administration or in situ vaccination therapies due to inflammatory toxicity. One approach to address this challenge involves better targeting the action of imidazoquinolines by caging them [...] Read more.
Despite broad anti-cancer efficacy as Toll-Like Receptor (TLR) 7/8 agonists, imidazoquinolines remain limited in use via systemic administration or in situ vaccination therapies due to inflammatory toxicity. One approach to address this challenge involves better targeting the action of imidazoquinolines by caging them as glycoconjugate prodrugs. Within cancer cells, imidazoquinoline glycoconjugates are activated by hydrolases prior to efflux by ABC transport proteins, where they then elicit tumoricidal effects from the assistance of bystander immune cells, such as tumor-infiltrating lymphocytes and associated macrophages, in local proximity. While this concept of Bystander-Assisted ImmunoTherapy (BAIT) has been established at a molecular level in vitro, tolerability or efficacy of BAIT has not been reported in vivo. Here, we evaluate the MTD and tumor growth delay efficacy of a lead BAIT prodrug (BAIT628) in a male C57BL/6 mouse TRAMP-C2 prostate cancer model to further establish this methodology. Overall, we find that systemic BAIT628 is well tolerated at over 5-fold the dose-limiting inflammatory toxicity of the parent imidazoquinoline (up to 5 mg/mouse/day I.P. for 10 days). Analyzing serum cytokines reveals that IL-10 production, elicited by the mannoside caging group, likely contributes to the enhanced MTD. Using BAIT628 as an in situ vaccination immunotherapy (seven times over 3 weeks) resulted in significant tumor growth delay and increased survival, both alone and in combination with a murinized α-PD-L1 checkpoint blockade. The tumor histology of tumor-infiltrating immune cell subsets (CD4+, CD8+, CD11c+) reveals significant increases in CD11c+ populations, consistent with TLR7/8 agonism. Overall, BAIT628 is well tolerated and exhibits significant efficacy in the TRAMP-C2 model. These results demonstrate how the BAIT approach can optimize imidazoquinolines for in vivo tolerability and subsequent efficacy as cancer immunotherapeutics. Full article
Show Figures

Figure 1

14 pages, 8033 KB  
Article
GSH-Responsive Nano-Photosensitizer for Potentiating Photodynamic Therapy Through Multi-Pronged Synergistic Upregulation of Ferroptosis Sensitivity
by Yunong Ma, Kexin Xu, Jing Feng, Xi Zhao, Peilin Tian, Jiayang Luo, Luyao Xu, Jiaxing Song and Cuixia Lu
Antioxidants 2025, 14(4), 407; https://doi.org/10.3390/antiox14040407 - 28 Mar 2025
Viewed by 1165
Abstract
Impeded by the limited light penetration of photodynamic therapy (PDT) to tissues and the hypoxic environment of solid tumors, the clinical therapeutic efficacy and application are below expectations. In this study, a glutathione (GSH)-responsive nano-photosensitizer, based on the chlorquinaldol (CQD)-loaded iron-containing nanorod composed [...] Read more.
Impeded by the limited light penetration of photodynamic therapy (PDT) to tissues and the hypoxic environment of solid tumors, the clinical therapeutic efficacy and application are below expectations. In this study, a glutathione (GSH)-responsive nano-photosensitizer, based on the chlorquinaldol (CQD)-loaded iron-containing nanorod composed of meso-tetra (4-carboxyphenyl) porphyrin (TCPP), was prepared to serve as the laser-ignited ferroptosis sensitizer to improve the tumoricidal effect of PDT. In the tumor microenvironment (TME) with elevated GSH levels, therapeutic cargos and ferrous ions are released and are accompanied by the degradation of the nano-photosensitizer and GSH exhaustion. This not only increases liable iron pool (LIP) accumulation by the released ferrous ions but also decreases glutathione peroxidase 4 (GPX4) activity by GSH exhaustion. Simultaneously, GSH exhaustion disrupts intracellular redox homeostasis, heightening NIR light irradiation-triggered photosensitive oxidative stress. Moreover, the released CQD elevates the level of intracellular reactive oxygen species (ROS), enabling the nanorods to gain an oxygen radical generation ability and enhancing the photosensitive oxidative therapeutic efficacy. Strikingly, CQD exacerbates the downregulation of GPX4 expression to promote the accumulation of lipid peroxides. Therefore, we herald a new paradigm for synergistically potentiating PDT based on the “all-in-one” nano-photosensitizer through the multi-pronged upregulation of ferroptosis sensitivity. Full article
(This article belongs to the Special Issue Nanotechnology and Redox Health)
Show Figures

Figure 1

22 pages, 5485 KB  
Article
Peptide-Conjugated Vascular Endothelial Extracellular Vesicles Encapsulating Vinorelbine for Lung Cancer Targeted Therapeutics
by Isha Gaurav, Abhimanyu Thakur, Kui Zhang, Sudha Thakur, Xin Hu, Zhijie Xu, Gaurav Kumar, Ravindran Jaganathan, Ashok Iyaswamy, Min Li, Ge Zhang and Zhijun Yang
Nanomaterials 2024, 14(20), 1669; https://doi.org/10.3390/nano14201669 - 17 Oct 2024
Cited by 6 | Viewed by 2374
Abstract
Lung cancer is one of the major cancer types and poses challenges in its treatment, including lack of specificity and harm to healthy cells. Nanoparticle-based drug delivery systems (NDDSs) show promise in overcoming these challenges. While conventional NDDSs have drawbacks, such as immune [...] Read more.
Lung cancer is one of the major cancer types and poses challenges in its treatment, including lack of specificity and harm to healthy cells. Nanoparticle-based drug delivery systems (NDDSs) show promise in overcoming these challenges. While conventional NDDSs have drawbacks, such as immune response and capture by the reticuloendothelial system (RES), extracellular vesicles (EVs) present a potential solution. EVs, which are naturally released from cells, can evade the RES without surface modification and with minimal toxicity to healthy cells. This makes them a promising candidate for developing a lung-cancer-targeting drug delivery system. EVs isolated from vascular endothelial cells, such as human umbilical endothelial-cell-derived EVs (HUVEC-EVs), have shown anti-angiogenic activity in a lung cancer mouse model; therefore, in this study, HUVEC-EVs were chosen as a carrier for drug delivery. To achieve lung-cancer-specific targeting, HUVEC-EVs were engineered to be decorated with GE11 peptides (GE11-HUVEC-EVs) via a postinsertional technique to target the epidermal growth factor receptor (EGFR) that is overexpressed on the surface of lung cancer cells. The GE11-HUVEC-EVs were loaded with vinorelbine (GE11-HUVEC-EVs-Vin), and then characterized and evaluated in in vitro and in vivo lung cancer models. Further, we examined the binding affinity of ABCB1, encoding P-glycoprotein, which plays a crucial role in chemoresistance via the efflux of the drug. Our results indicate that GE11-HUVEC-EVs-Vin effectively showed tumoricidal effects against cell and mouse models of lung cancer. Full article
Show Figures

Figure 1

13 pages, 1817 KB  
Article
Tumoricidal Activity and Side Effects of Radiolabeled Anti-NCAM [131I]-Iodine-ERIC1 in Neuroblastoma-Bearing Mice
by Thomas Fischer, Felix Dietlein, Detlev Bongartz, Martin Klehr, Beate Zimmermanns, Matthias Schmidt, Angela Mohr, Fabian Mohr, Ferdinand Sudbrock, Philipp Krapf, Alexander Drzezga, Markus Dietlein and Klaus Schomäcker
Int. J. Mol. Sci. 2024, 25(19), 10737; https://doi.org/10.3390/ijms251910737 - 5 Oct 2024
Cited by 1 | Viewed by 1623
Abstract
Preliminary studies on a radioactive antibody against the neural cell adhesion molecule (NCAM) demonstrated a significant accumulation of [131I]I-ERIC1 in neuroblastoma tumor cells in mice. This study aims to validate the therapeutic efficacy and potential adverse effects of these radioactive immunoconjugates [...] Read more.
Preliminary studies on a radioactive antibody against the neural cell adhesion molecule (NCAM) demonstrated a significant accumulation of [131I]I-ERIC1 in neuroblastoma tumor cells in mice. This study aims to validate the therapeutic efficacy and potential adverse effects of these radioactive immunoconjugates (RICs) in neuroblastoma-bearing mice. To determine the highest tolerated dose, healthy SCID mice received 1 to 22 MBq of [131I]I-ERIC1, with the survival time measured. Tumor response was evaluated by administering 0.8 to 22 MBq of [131I]I-ERIC1 to neuroblastoma-bearing mice and assessing tumor size and systemic toxicity through body weight, blood counts, and survival. It was observed that doses up to approximately 3 MBq per animal (150 MBq/kg) were well tolerated, whereas higher doses resulted in systemic toxicity and death. The neuroblastomas exhibited a dose-dependent response, with optimal therapeutic efficacy achieved at 1.8–2.5 MBq per animal (90–125 MBq/kg), significantly extending survival by a factor of five. The antibody ERIC1 is a promising vehicle for the transport of beta emitters into NCAM-positive tumor tissue. An optimal dosage of the [131I]I-ERIC1 antibody can be established with a balance of tumor-static effects and adverse effects, resulting in a marked extension of survival time. Full article
Show Figures

Figure 1

9 pages, 1845 KB  
Article
All-Natural Gelatin-Based Nanoemulsion Loaded with TLR 7/8 Agonist for Efficient Modulation of Macrophage Polarization for Immunotherapy
by Ritabrita Goswami, Ahmed Nabawy, Mingdi Jiang, Yagiz Anil Cicek, Muhammad Aamir Hassan, Harini Nagaraj, Xianzhi Zhang and Vincent M Rotello
Nanomaterials 2024, 14(19), 1556; https://doi.org/10.3390/nano14191556 - 26 Sep 2024
Cited by 4 | Viewed by 2238
Abstract
Macrophages are multifunctional immune cells essential for both innate and adaptive immune responses. Tumor-associated macrophages (TAMs) often adopt a tumor-promoting M2-like phenotype, aiding tumor progression and immune evasion. Reprogramming TAMs to a tumoricidal M1-like phenotype is an emerging target for cancer immunotherapy. Resiquimod, [...] Read more.
Macrophages are multifunctional immune cells essential for both innate and adaptive immune responses. Tumor-associated macrophages (TAMs) often adopt a tumor-promoting M2-like phenotype, aiding tumor progression and immune evasion. Reprogramming TAMs to a tumoricidal M1-like phenotype is an emerging target for cancer immunotherapy. Resiquimod, a TLR7/8 agonist, can repolarize macrophages from the M2- to M1-like phenotype but is limited by poor solubility. We developed a gelatin nanoemulsion for the loading and delivery of resiquimod, utilizing eugenol oil as the liquid phase and riboflavin-crosslinked gelatin as a scaffold. These nanoemulsions showed high stability, low toxicity, and effective macrophage repolarization, significantly enhancing pro-inflammatory markers and anticancer activity in co-culture models. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Figure 1

20 pages, 2486 KB  
Review
Natural Killer-Based Therapy: A Prospective Thought for Cancer Treatment Related to Diversified Drug Delivery Pathways
by Jing Zang, Yijun Mei, Shiguo Zhu, Shaoping Yin, Nianping Feng, Tianyuan Ci and Yaqi Lyu
Pharmaceutics 2024, 16(7), 939; https://doi.org/10.3390/pharmaceutics16070939 - 14 Jul 2024
Cited by 2 | Viewed by 2438
Abstract
Immunotherapy has been a research hotspot due to its low side effects, long-lasting efficacy, and wide anti-tumor spectrum. Recently, NK cell-based immunotherapy has gained broad attention for its unique immunological character of tumor identification and eradication and low risk of graft-versus-host disease and [...] Read more.
Immunotherapy has been a research hotspot due to its low side effects, long-lasting efficacy, and wide anti-tumor spectrum. Recently, NK cell-based immunotherapy has gained broad attention for its unique immunological character of tumor identification and eradication and low risk of graft-versus-host disease and cytokine storm. With the cooperation of a drug delivery system (DDS), NK cells activate tumoricidal activity by adjusting the balance of the activating and inhibitory signals on their surface after drug-loaded DDS administration. Moreover, NK cells or NK-derived exosomes can also be applied as drug carriers for distinct modification to promote NK activation and exert anti-tumor effects. In this review, we first introduce the source and classification of NK cells and describe the common activating and inhibitory receptors on their surface. Then, we summarize the strategies for activating NK cells in vivo through various DDSs. Finally, the application prospects of NK cells in tumor immunotherapy are also discussed. Full article
(This article belongs to the Special Issue Advanced Nanopharmaceuticals for Anticancer Therapy)
Show Figures

Figure 1

31 pages, 5790 KB  
Article
Chlorin Conjugates in Photodynamic Chemotherapy for Triple-Negative Breast Cancer
by Meden F. Isaac-Lam
Pharmaceuticals 2024, 17(5), 576; https://doi.org/10.3390/ph17050576 - 30 Apr 2024
Cited by 3 | Viewed by 2540
Abstract
Breast cancer (BC) is the most common type of cancer in women and the number of new cases in the US is still increasing each year. Triple-negative breast cancer (TNBC), which comprises 15–20% of all breast cancer, is a heterogeneous disease and is [...] Read more.
Breast cancer (BC) is the most common type of cancer in women and the number of new cases in the US is still increasing each year. Triple-negative breast cancer (TNBC), which comprises 15–20% of all breast cancer, is a heterogeneous disease and is considered the most aggressive type of breast cancer due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expressions for treatments. Traditional chemotherapy is the standard protocol for the treatment of TNBC. Toxicity and multidrug resistance are major drawbacks to chemotherapy. The lack of molecular targets and poor prognosis for TNBC prompts an urgent need to discover novel therapeutic strategies to improve clinical outcomes and quality of life for patients. Photodynamic therapy (PDT) or light treatment is a binary anti-cancer procedure that uses a photosensitizer (PS) that, upon light activation, produces cytotoxic oxygen species, destroying tumor cells. PDT is minimally invasive and can be repeated a few times without accumulating significant toxicity in the surrounding tissues. The primary goal of this study was to investigate in vitro photodynamic chemotherapy as a ternary combination therapy using our synthesized photosensitizers (chlorin–vitamin conjugates and their corresponding indium complexes) co-treated with known chemotherapeutic agents (taxol, doxorubicin, cisplatin, fluorouracil, or methotrexate) in the presence of light and determine the optimum conditions as a pre-clinical study of an enhanced tumoricidal effect against TNBC. Our results indicated that the best combination for an effective chemophotodynamic effect involves a ternary treatment of the indium complex of the chlorin–lipoic acid conjugate (InCLA) co-treated with taxol, which exhibited strong synergism at the nanomolar concentration when combined in the presence of visible light irradiation. Other ternary combinations containing taxol with a synergistic anti-tumor effect against TNBC include chlorin–pantothenic acid (CPA) and chlorin–biotin (CBTN) conjugates. Several other ternary combinations containing InCLA, CBTN, and CPA with either cisplatin, fluorouracil, or methotrexate were identified to generate a synergistic or additive effect. The light dosage remained constant, but the dosages of photosensitizers and chemotherapy drugs were varied to obtain the lowest possible concentration for the desired effect. The synergistic, additive or antagonistic effects of the drug combinations were determined based on the Chou–Talalay method, with InCLA–taxol having the lowest combination index (CI) of 0.25. Fluorescence and transmission electron microscopy (TEM) images provided evidence of apoptosis as the preferred mode of cell death. Our study demonstrated the combination of PDT and chemotherapy as a potential treatment option for TNBC patients. Full article
(This article belongs to the Special Issue Photodynamic Therapy 2023)
Show Figures

Figure 1

15 pages, 2462 KB  
Article
(Pentamethylcyclopentadienyl)chloridoiridium(III) Complex Bearing Bidentate Ph2PCH2CH2SPh-κPS Ligand
by Gerd Ludwig, Ivan Ranđelović, Dušan Dimić, Teodora Komazec, Danijela Maksimović-Ivanić, Sanja Mijatović, Tobias Rüffer and Goran N. Kaluđerović
Biomolecules 2024, 14(4), 420; https://doi.org/10.3390/biom14040420 - 30 Mar 2024
Cited by 5 | Viewed by 2285
Abstract
The (pentamethylcyclopentadienyl)chloridoiridium(III) complex bearing a κPS-bonded Ph2PCH2CH2SPh ligand ([Ir(η5-C5Me5)Cl(Ph2P(CH2)2SPh-κP,κS)]PF6, (1)] was synthesized and [...] Read more.
The (pentamethylcyclopentadienyl)chloridoiridium(III) complex bearing a κPS-bonded Ph2PCH2CH2SPh ligand ([Ir(η5-C5Me5)Cl(Ph2P(CH2)2SPh-κP,κS)]PF6, (1)] was synthesized and characterized. Multinuclear (1H, 13C and 31P) NMR spectroscopy was employed for the determination of the structure. Moreover, SC-XRD confirmed the proposed structure belongs to the “piano stool” type. The Hirshfeld surface analysis outlined the most important intermolecular interactions in the structure. The crystallographic structure was optimized at the B3LYP-D3BJ/6-311++G(d,p)(H,C,P,S,Cl)/LanL2DZ(Ir) level of theory. The applicability of this level was verified through a comparison of experimental and theoretical bond lengths and angles, and 1H and 13C NMR chemical shifts. The Natural Bond Orbital theory was used to identify and quantify the intramolecular stabilization interactions, especially those between donor atoms and Ir(III) ions. Complex 1 was tested on antitumor activity against five human tumor cell lines: MCF-7 breast adenocarcinoma, SW480 colon adenocarcinoma, 518A2 melanoma, 8505C human thyroid carcinoma and A253 submandibular carcinoma. Complex 1 showed superior antitumor activity against cisplatin-resistant MCF-7, SW480 and 8505C cell lines. The mechanism of tumoricidal action on 8505C cells indicates the involvement of caspase-induced apoptosis, accompanied by a considerable reduction in ROS/RNS and proliferation potential of treated cells. Full article
Show Figures

Figure 1

43 pages, 48297 KB  
Article
Novel Antineoplastic Inducers of Mitochondrial Apoptosis in Human Cancer Cells
by Andreas J. Kesel
Molecules 2024, 29(4), 914; https://doi.org/10.3390/molecules29040914 - 19 Feb 2024
Viewed by 3999
Abstract
I propose a new strategy to suppress human cancer completely with two entirely new drug compounds exploiting cancer’s Warburg effect characterized by a defective mitochondrial aerobic respiration, substituted by cytosolic aerobic fermentation/glycolysis of D-(+)-glucose into L-(+)-lactic acid. The two essentially new drugs, compound [...] Read more.
I propose a new strategy to suppress human cancer completely with two entirely new drug compounds exploiting cancer’s Warburg effect characterized by a defective mitochondrial aerobic respiration, substituted by cytosolic aerobic fermentation/glycolysis of D-(+)-glucose into L-(+)-lactic acid. The two essentially new drugs, compound 1 [P(op)T(est)162] and compound 3 (PT167), represent new highly symmetric, four-bladed propeller-shaped polyammonium cations. The in vitro antineoplastic highly efficacious drug compound 3 represents a covalent combination of compound 1 and compound 2 (PT166). The intermediate drug compound 2 is an entirely new colchic(in)oid derivative synthesized from colchicine. Compound 2’s structure was determined using X-ray crystallography. Compound 1 and compound 3 were active in vitro versus 60 human cancer cell lines of the National Cancer Institute (NCI) Developmental Therapeutics Program (DTP) 60-cancer cell testing. Compound 1 and compound 3 not only stop the growth of cancer cells to ±0% (cancerostatic effect) but completely kill nearly all 60 cancer cells to a level of almost −100% (tumoricidal effect). Compound 1 and compound 3 induce mitochondrial apoptosis (under cytochrome c release) in all cancer cells tested by (re)activating (in most cancers impaired) p53 function, which results in a decrease in cancer’s dysregulated cyclin D1 and an induction of the cell cycle-halting cyclin-dependent kinase inhibitor p21Waf1/p21Cip1. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

17 pages, 2437 KB  
Communication
Transcriptional Targeting of Dendritic Cells Using an Optimized Human Fascin1 Gene Promoter
by Yanira Zeyn, Dominika Hobernik, Ulrich Wilk, Jana Pöhmerer, Christoph Hieber, Carolina Medina-Montano, Nadine Röhrig, Caroline F. Strähle, Andrea K. Thoma-Kress, Ernst Wagner, Matthias Bros and Simone Berger
Int. J. Mol. Sci. 2023, 24(23), 16938; https://doi.org/10.3390/ijms242316938 - 29 Nov 2023
Cited by 1 | Viewed by 2552
Abstract
Deeper knowledge about the role of the tumor microenvironment (TME) in cancer development and progression has resulted in new strategies such as gene-based cancer immunotherapy. Whereas some approaches focus on the expression of tumoricidal genes within the TME, DNA-based vaccines are intended to [...] Read more.
Deeper knowledge about the role of the tumor microenvironment (TME) in cancer development and progression has resulted in new strategies such as gene-based cancer immunotherapy. Whereas some approaches focus on the expression of tumoricidal genes within the TME, DNA-based vaccines are intended to be expressed in antigen-presenting cells (e.g., dendritic cells, DCs) in secondary lymphoid organs, which in turn induce anti-tumor T cell responses. Besides effective delivery systems and the requirement of appropriate adjuvants, DNA vaccines themselves need to be optimized regarding efficacy and selectivity. In this work, the concept of DC-focused transcriptional targeting was tested by applying a plasmid encoding for the luciferase reporter gene under the control of a derivative of the human fascin1 gene promoter (pFscnLuc), comprising the proximal core promoter fused to the normally more distantly located DC enhancer region. DC-focused activity of this reporter construct was confirmed in cell culture in comparison to a standard reporter vector encoding for luciferase under the control of the strong ubiquitously active cytomegalovirus promoter and enhancer (pCMVLuc). Both plasmids were also compared upon intravenous administration in mice. The organ- and cell type-specific expression profile of pFscnLuc versus pCMVLuc demonstrated favorable activity especially in the spleen as a central immune organ and within the spleen in DCs. Full article
(This article belongs to the Special Issue Advances in Vaccines, Adjuvants and Delivery Technologies)
Show Figures

Graphical abstract

Back to TopTop