Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (64)

Search Parameters:
Keywords = intra-tumor hypoxia

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 3455 KiB  
Review
Recent Advances in Nanoparticle and Nanocomposite-Based Photodynamic Therapy for Cervical Cancer: A Review
by Dorota Bartusik-Aebisher, Mohammad A. Saad, Agnieszka Przygórzewska and David Aebisher
Cancers 2025, 17(15), 2572; https://doi.org/10.3390/cancers17152572 - 4 Aug 2025
Abstract
Cervical cancer represents a significant global health challenge. Photodynamic therapy (PDT) appears to be a promising, minimally invasive alternative to standard treatments. However, the clinical efficacy of PDT is sometimes limited by the low solubility and aggregation of photosensitizers, their non-selective distribution in [...] Read more.
Cervical cancer represents a significant global health challenge. Photodynamic therapy (PDT) appears to be a promising, minimally invasive alternative to standard treatments. However, the clinical efficacy of PDT is sometimes limited by the low solubility and aggregation of photosensitizers, their non-selective distribution in the body, hypoxia in the tumor microenvironment, and limited light penetration. Recent advances in nanoparticle and nanocomposite platforms have addressed these challenges by integrating multiple functional components into a single delivery system. By encapsulating or conjugating photosensitizers in biodegradable matrices, such as mesoporous silica, organometallic structures and core–shell construct nanocarriers increase stability in water and extend circulation time, enabling both passive and active targeting through ligand decoration. Up-conversion and dual-wavelength responsive cores facilitate deep light conversion in tissues, while simultaneous delivery of hypoxia-modulating agents alleviates oxygen deprivation to sustain reactive oxygen species generation. Controllable “motor-cargo” constructs and surface modifications improve intratumoral diffusion, while aggregation-induced emission dyes and plasmonic elements support real-time imaging and quantitative monitoring of therapeutic response. Together, these multifunctional nanosystems have demonstrated potent cytotoxicity in vitro and significant tumor suppression in vivo in mouse models of cervical cancer. Combining targeted delivery, controlled release, hypoxia mitigation, and image guidance, engineered nanoparticles provide a versatile and powerful platform to overcome the current limitations of PDT and pave the way toward more effective, patient-specific treatments for cervical malignancies. Our review of the literature summarizes studies on nanoparticles and nanocomposites used in PDT monotherapy for cervical cancer, published between 2023 and July 2025. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

28 pages, 2091 KiB  
Review
Spatiotemporal Heterogeneity of Tumor Glucose Metabolism Reprogramming: From Single-Cell Mechanisms to Precision Interventions
by Xiaoxue Chai, Qian Tao and Lili Li
Int. J. Mol. Sci. 2025, 26(14), 6901; https://doi.org/10.3390/ijms26146901 - 18 Jul 2025
Viewed by 583
Abstract
Glucose metabolism reprogramming as a defining hallmark of cancer has become a pivotal frontier in oncology research. Recent technological advances in single-cell sequencing, spatial omics, and metabolic imaging have transformed the field from static bulk analyses to dynamic investigations of spatiotemporal heterogeneity at [...] Read more.
Glucose metabolism reprogramming as a defining hallmark of cancer has become a pivotal frontier in oncology research. Recent technological advances in single-cell sequencing, spatial omics, and metabolic imaging have transformed the field from static bulk analyses to dynamic investigations of spatiotemporal heterogeneity at a single-cell resolution. This review systematically summarizes the current knowledge on tumor glucose metabolism dynamics, discussing spatial heterogeneity and temporal evolution patterns, metabolic subpopulation interactions revealed by single-cell metabolomics, the glucose metabolism–epigenetics–immunology regulatory axis, and therapeutic strategies targeting metabolic vulnerabilities. Recent technological advances in single-cell sequencing and spatial omics have transformed our understanding of tumor glucose metabolism by providing high-resolution insights into metabolic heterogeneity and regulatory mechanisms, contrasting with classical bulk analyses. Spatiotemporal heterogeneity critically influences therapeutic outcomes by enabling tumor cells to adapt metabolically under selective pressures (e.g., hypoxia, nutrient deprivation), fostering treatment resistance and relapse. Deciphering these dynamics is essential for developing spatiotemporally targeted strategies that address intratumoral diversity and microenvironmental fluctuations. By integrating cutting-edge advances, this review deepens our understanding of tumor metabolic complexity and provides a conceptual framework for developing spatiotemporally precise interventions. Full article
Show Figures

Figure 1

18 pages, 4058 KiB  
Article
Evaluating Immune Activation Feasibility in Pancreatic Ductal Adenocarcinoma via Oxygen Bubble-Induced Anti-Vascular Therapy
by Tzu-Yun Chiu, Yi-Jia Zho and Yi-Ju Ho
Pharmaceutics 2025, 17(5), 645; https://doi.org/10.3390/pharmaceutics17050645 - 13 May 2025
Viewed by 473
Abstract
Background/Objectives: Anti-vascular therapy presents a potential strategy for activating anti-tumor immunity. Disrupted vascular debris provides effective antigens that activate dendritic cells, leading to subsequent immune responses. However, the resulting tumor hypoxia following vascular disruption may contribute to immune suppression, thereby hindering effective immune [...] Read more.
Background/Objectives: Anti-vascular therapy presents a potential strategy for activating anti-tumor immunity. Disrupted vascular debris provides effective antigens that activate dendritic cells, leading to subsequent immune responses. However, the resulting tumor hypoxia following vascular disruption may contribute to immune suppression, thereby hindering effective immune activation. Ultrasound-stimulated microbubble cavitation can locally disrupt tumor vessels through mechanical effects to achieve physical anti-vascular therapy. Therefore, this study designed oxygen-loaded nanobubbles (ONBs) to combine anti-vascular effects with local oxygen release under ultrasound stimulation. The feasibility of enhancing anti-tumor immune activation by alleviating tumor hypoxia was evaluated. Methods: A murine pancreatic subcutaneous solid tumor model was used to evaluate the efficacy of anti-vascular therapy-associated immunotherapy. Results: After ONB treatment, tumor perfusion was reduced to 52 ± 5%, which resulted in a subsequent 57 ± 11% necrosis and a 29 ± 4% reduction in hypoxia, demonstrating the anti-vascular effect and reoxygenation, respectively. However, subsequent immune responses exhibited no significant activation in intratumoral cytokine expression or splenic immune cell composition. Primary tumors exhibited a 15.7 ± 5.0% increase in necrosis following ONB treatment, but distant tumor growth was not significantly inhibited. Conclusions: These results highlighted a crucial issue regarding the complex correlations between vessel disruption, antigen production, oxygen delivery, hypoxia, and immunity when combining anti-vascular therapy with immunotherapy. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

14 pages, 672 KiB  
Review
Tumor Hypoxia: How Conventional Histology Is Reshaped in Breast Carcinoma
by Péter Juhász and Gábor Méhes
Int. J. Mol. Sci. 2025, 26(9), 4423; https://doi.org/10.3390/ijms26094423 - 6 May 2025
Viewed by 562
Abstract
Intratumoral hypoxia is common in any form of malignancy initializing focal necrosis or tumor cell adaptation. Hypoxia inducible factor-1-driven reprogramming favors the loss of tumor cell proliferation (quiescence) and partial cellular reversion, induces stemness and/or mesenchymal-like features in the exposed tumor areas. The [...] Read more.
Intratumoral hypoxia is common in any form of malignancy initializing focal necrosis or tumor cell adaptation. Hypoxia inducible factor-1-driven reprogramming favors the loss of tumor cell proliferation (quiescence) and partial cellular reversion, induces stemness and/or mesenchymal-like features in the exposed tumor areas. The characteristic hypoxia-driven tumor cell phenotype is principally directed to reduce energy consumption and to enhance survival, but the gained features also contribute to growth advantage and induce the reorganization of the microenvironment and protective mechanisms against external stress. The hypoxia-induced phenotypic changes are at least in part reflected by conventional morphology in breast carcinoma. Intratumoral variability of classical morphological signs, such as the growth pattern, the histological grade, cell proliferation, necrosis, microcalcification, angiogenesis, and the immune cell infiltration is also related with the co-existence of hypoxic areas. Thus, a deeper understanding of hypoxia-activated mechanisms is required. The current paper aims to summarize the major tissue factors involved in the response to hypoxia and their potential contribution to the breast carcinoma phenotype. Full article
(This article belongs to the Special Issue Biomarkers of Tumor Progression, Prognosis and Therapy: 2nd Edition)
Show Figures

Figure 1

31 pages, 2559 KiB  
Review
Roles of Post-Translational Modifications of Transcription Factors Involved in Breast Cancer Hypoxia
by Logan Seymour, Niyogushima Nuru, Kaya R. Johnson, Jennifer Michel Villalpando Gutierrez, Victor Tochukwu Njoku, Costel C. Darie and Anca-Narcisa Neagu
Molecules 2025, 30(3), 645; https://doi.org/10.3390/molecules30030645 - 1 Feb 2025
Cited by 2 | Viewed by 2359
Abstract
BC is the most commonly diagnosed cancer and the second leading cause of cancer death among women worldwide. Cellular stress is a condition that leads to disrupted homeostasis by extrinsic and intrinsic factors. Among other stressors, hypoxia is a driving force for breast [...] Read more.
BC is the most commonly diagnosed cancer and the second leading cause of cancer death among women worldwide. Cellular stress is a condition that leads to disrupted homeostasis by extrinsic and intrinsic factors. Among other stressors, hypoxia is a driving force for breast cancer (BC) progression and a general hallmark of solid tumors. Thus, intratumoral hypoxia is an important determinant of invasion, metastasis, treatment failure, prognosis, and patient mortality. Acquisition of the epithelial–mesenchymal transition (EMT) phenotype is also a consequence of tumor hypoxia. The cellular response to hypoxia is mainly regulated by the hypoxia signaling pathway, governed by hypoxia-inducible factors (HIFs), mainly HIF1α. HIFs are a family of transcription factors (TFs), which induce the expression of target genes involved in cell survival and proliferation, metabolic reprogramming, angiogenesis, resisting apoptosis, invasion, and metastasis. HIF1α cooperates with a large number of other TFs. In this review, we focused on the crosstalk and cooperation between HIF1α and other TFs involved in the cellular response to hypoxia in BC. We identified a cluster of TFs, proposed as the HIF1α-TF interactome, that orchestrates the transcription of target genes involved in hypoxia, due to their post-translational modifications (PTMs), including phosphorylation/dephosphorylation, ubiquitination/deubiquitination, SUMOylation, hydroxylation, acetylation, S-nitrosylation, and palmitoylation. PTMs of these HIF1α-related TFs drive their stability and activity, degradation and turnover, and the bidirectional translocation between the cytoplasm or plasma membrane and nucleus of BC cells, as well as the transcription/activation of proteins encoded by oncogenes or inactivation of tumor suppressor target genes. Consequently, PTMs of TFs in the HIF1α interactome are crucial regulatory mechanisms that drive the cellular response to oxygen deprivation in BC cells. Full article
(This article belongs to the Special Issue Featured Review Papers in Bioorganic Chemistry)
Show Figures

Figure 1

13 pages, 3335 KiB  
Article
Prognostic Significance of Microvessel Density and Hypoxic Markers in Canine Osteosarcoma: Insights into Angiogenesis and Tumor Aggressiveness
by Cecilia Gola, Marcella Massimini, Emanuela Morello, Lorella Maniscalco, Luiza Cesar Conti, Mariarita Romanucci, Matteo Olimpo, Leonardo Della Salda and Raffaella De Maria
Animals 2024, 14(22), 3181; https://doi.org/10.3390/ani14223181 - 6 Nov 2024
Viewed by 1476
Abstract
Canine osteosarcoma (OSA) is an aggressive and highly malignant tumor of bone with a poor prognosis and it mirrors the disease in humans. Angiogenesis, the formation of new blood vessels, is driven by hypoxia-induced factors such as HIF-1α and VEGF, both of which [...] Read more.
Canine osteosarcoma (OSA) is an aggressive and highly malignant tumor of bone with a poor prognosis and it mirrors the disease in humans. Angiogenesis, the formation of new blood vessels, is driven by hypoxia-induced factors such as HIF-1α and VEGF, both of which play a crucial role in tumor growth and metastasis. However, the role of angiogenesis in OSA remains a topic of ongoing debate. This study aimed to investigate the relationship between angiogenesis, measured by intratumoral microvessel density (MVD), hypoxic markers, and clinical outcomes in 28 dogs diagnosed with appendicular OSA. Clinicopathological data such as age, breed distribution, tumor localization, histopathological subtypes, and metastatic behavior were consistent with reported epidemiologic characteristics of canine OSA, though no significant correlation was found among these variables. The results indicated a significant association between higher MVD and high-grade OSA (p = 0.029), suggesting that increased tumor vascularization is linked to more aggressive tumor behavior. Additionally, elevated VEGF expression was strongly correlated with disease-free interval DFI), with a p-value of 0.045. Although HIF-1α positivity showed a trend towards poorer survival, the results did not reach statistical significance (p = 0.07). These findings highlight the potential role of VEGF as a valuable prognostic marker in canine OSA, which could have potentially important implications for therapeutic targeting and clinical management of the disease. This study advances the understanding of angiogenesis in canine OSA, while emphasizing the need for continued research into the complex mechanisms regulating the interplay between hypoxia, angiogenesis and tumor progression. Full article
(This article belongs to the Section Companion Animals)
Show Figures

Figure 1

18 pages, 1983 KiB  
Article
An Exogenous Ketone Ester Slows Tumor Progression in Murine Breast and Renal Cancer Models
by Henry Nnaemeka Ogbonna, Zachary Roberts, Nicholas Godwin, Pia Muri, William J. Turbitt, Zoey N. Swalley, Francesca R. Dempsey, Holly R. Stephens, Jianqing Zhang, Eric P. Plaisance and Lyse A. Norian
Cancers 2024, 16(19), 3390; https://doi.org/10.3390/cancers16193390 - 4 Oct 2024
Cited by 1 | Viewed by 3022
Abstract
Background/Objectives: Ketone esters (KEs) exhibit promise as anti-cancer agents but their impact on spontaneous metastases remains poorly understood. Although consumption of a ketogenic diet (KD) that is low in carbohydrates and high in fats can lead to KE production in vivo, the restrictive [...] Read more.
Background/Objectives: Ketone esters (KEs) exhibit promise as anti-cancer agents but their impact on spontaneous metastases remains poorly understood. Although consumption of a ketogenic diet (KD) that is low in carbohydrates and high in fats can lead to KE production in vivo, the restrictive composition of KDs may diminish adherence in cancer patients. Methods: We investigated the effects of an exogenous ketone ester-supplemented (eKET), carbohydrate-replete diet on tumor growth, metastasis, and underlying mechanisms in orthotopic models of metastatic breast (4T1-Luc) and renal (Renca-Luc) carcinomas. Mice were randomized to diet after tumor challenge. Results: Administration of KEs did not alter tumor cell growth in vitro. However, in mice, our eKET diet increased circulating β-hydroxybutyrate and inhibited primary tumor growth and lung metastasis in both models. Body composition analysis illustrated the overall safety of eKET diet use, although it was associated with a loss of fat mass in mice with renal tumors. Immunogenetic profiling revealed divergent intratumoral eKET-related changes by tumor type. In mammary tumors, Wnt and TGFβ pathways were downregulated, whereas in renal tumors, genes related to hypoxia and DNA damage repair were downregulated. Conclusions: Thus, our eKET diet exerts potent antitumor and antimetastatic effects in both breast and renal cancer models, albeit with different modes of action and physiologic effects. Its potential as an adjuvant dietary approach for patients with diverse cancer types should be explored further. Full article
(This article belongs to the Section Methods and Technologies Development)
Show Figures

Figure 1

23 pages, 9906 KiB  
Article
In Vitro Cytotoxic Potential and In Vivo Antitumor Effects of NOS/PDK-Inhibitor T1084
by Marina Filimonova, Anna Shitova, Ljudmila Shevchenko, Olga Soldatova, Valentina Surinova, Vitaly Rybachuk, Alexander Kosachenko, Kirill Nikolaev, Irina Volkova, Grigory Demyashkin, Tatjana P. Stanojkovic, Zeljko Zizak, Sergey Ivanov, Petr Shegay, Andrey Kaprin and Alexander Filimonov
Int. J. Mol. Sci. 2024, 25(17), 9711; https://doi.org/10.3390/ijms25179711 - 8 Sep 2024
Viewed by 1497
Abstract
Previously, we showed the antitumor activity of the new NOS/PDK inhibitor T1084 (1-isobutanoyl-2-isopropylisothiourea dichloroacetate). The present study included an assessment of in vitro cytotoxicity against human malignant and normal cells according to the MTT-test and in vivo antitumor effects in solid tumor models [...] Read more.
Previously, we showed the antitumor activity of the new NOS/PDK inhibitor T1084 (1-isobutanoyl-2-isopropylisothiourea dichloroacetate). The present study included an assessment of in vitro cytotoxicity against human malignant and normal cells according to the MTT-test and in vivo antitumor effects in solid tumor models in comparison with precursor compounds T1023 (NOS inhibitor; 1-isobutanoyl-2-isopropylisothiourea hydrobromide) and Na-DCA (PDK inhibitor; sodium dichloroacetate), using morphological, histological, and immunohistochemical methods. The effects of T1084 and T1023 on the in vitro survival of normal (MRC-5) and most malignant cells (A375, MFC-7, K562, OAW42, and PC-3) were similar and quantitatively equal. At the same time, melanoma A375 cells showed 2–2.5 times higher sensitivity (IC50: 0.39–0.41 mM) to the cytotoxicity of T1023 and T1084 than other cells. And only HeLa cells showed significantly higher sensitivity to the cytotoxicity of T1084 compared to T1023 (IC50: 0.54 ± 0.03 and 0.81 ± 0.02 mM). Comparative studies of the in vivo antitumor effects of Na-DCA, T1023, and T1084 on CC-5 cervical cancer and B-16 melanoma in mice were conducted with subchronic daily i.p. administration of these agents at an equimolar dose of 0.22 mmol/kg (33.6, 60.0, and 70.7 mg/kg, respectively). Cervical cancer CC-5 fairly quickly evaded the effects of both Na-DCA and T1023. So, from the end of the first week of Na-DCA or T1023 treatment, the tumor growth inhibition (TGI) began to decrease from 40% to an insignificant level by the end of the observation. In contrast, in two independent experiments, CC-5 showed consistently high sensitivity to the action of T1084: a significant antitumor effect with high TGI (43–58%) was registered throughout the observation, without any signs of neoplasia adaptation. The effect of precursor compounds on melanoma B-16 was either minimal (for Na-DCA) or moderate (for T1023) with TGI only 33%, which subsequently decreased by the end of the experiment. In contrast, the effect of T1084 on B-16 was qualitatively more pronounced and steadily increasing; it was accompanied by a 3-fold expansion of necrosis and dystrophy areas, a decrease in proliferation, and increased apoptosis of tumor cells. Morphologically, the T1084 effect was 2-fold superior to the effects of T1023—the TGI index reached 59–62%. This study suggests that the antitumor effects of T1084 develop through the interaction of NOS-dependent and PDK-dependent pathophysiological effects of this NOS/PDK inhibitor. The NOS inhibitory activity of T1084 exerts an anti-angiogenic effect on neoplasia. At the same time, the PDK inhibitory activity of T1084 enhances the cytotoxicity of induced intratumoral hypoxia and suppresses the development of neoplasia adaptation to anti-angiogenic stress. Such properties allow T1084 to overcome tumor resistance and realize a stable synergistic antitumor effect. Full article
Show Figures

Figure 1

15 pages, 2026 KiB  
Article
Hypoxia-Targeted Immunotherapy with PD-1 Blockade in Head and Neck Cancer
by Risa Wakisaka, Hidekiyo Yamaki, Michihisa Kono, Takahiro Inoue, Ryosuke Sato, Hiroki Komatsuda, Kenzo Ohara, Akemi Kosaka, Takayuki Ohkuri, Toshihiro Nagato, Kan Kishibe, Koh Nakayama, Hiroya Kobayashi, Takumi Kumai and Miki Takahara
Cancers 2024, 16(17), 3013; https://doi.org/10.3390/cancers16173013 - 29 Aug 2024
Viewed by 1488
Abstract
Intratumoral hypoxia is associated with tumor progression, aggressiveness, and therapeutic resistance in several cancers. Hypoxia causes cancer cells to experience replication stress, thereby activating DNA damage and repair pathways. MutT homologue-1 (MTH1, also known as NUDT1), a member of the Nudix family, maintains [...] Read more.
Intratumoral hypoxia is associated with tumor progression, aggressiveness, and therapeutic resistance in several cancers. Hypoxia causes cancer cells to experience replication stress, thereby activating DNA damage and repair pathways. MutT homologue-1 (MTH1, also known as NUDT1), a member of the Nudix family, maintains the genomic integrity and viability of tumor cells in the hypoxic tumor microenvironment. Although hypoxia is associated with poor prognosis and can cause therapeutic resistance by regulating the microenvironment, it has not been considered a treatable target in cancer. This study aimed to investigate whether hypoxia-induced MTH1 is a useful target for immunotherapy and whether hypoxic conditions influence the antitumor activity of immune cells. Our results showed that MTH1 expression was elevated under hypoxic conditions in head and neck cancer cell lines. Furthermore, we identified a novel MTH1-targeting epitope peptide that can activate peptide-specific CD4+ helper T cells with cytotoxic activity. The proliferation and cytotoxic activity of T cells were maintained under hypoxic conditions, and PD-1 blockade further augmented the cytotoxicity. These results indicate that MTH1-targeted immunotherapy combined with checkpoint blockade can be an effective strategy for the treatment of hypoxic tumors. Full article
Show Figures

Figure 1

19 pages, 17538 KiB  
Article
A Platelet-Powered Drug Delivery System for Enhancing Chemotherapy Efficacy for Liver Cancer Using the Trojan Horse Strategy
by Hao Huang, Xiaoping Wang, Ziqing Gao, Hongyi Bao, Xiaopeng Yuan, Chao Chen, Donglin Xia and Xiangqian Wang
Pharmaceutics 2024, 16(7), 905; https://doi.org/10.3390/pharmaceutics16070905 - 5 Jul 2024
Viewed by 1571
Abstract
Optimizing the delivery and penetration of nano-sized drugs within liver cancer sites, along with remodeling the tumor microenvironment, is crucial for enhancing the efficacy of chemotherapeutic agents. For this study, a platelet (PLT)-mediated nanodrug delivery system (DASA+ATO@PLT) was developed to improve the effectiveness [...] Read more.
Optimizing the delivery and penetration of nano-sized drugs within liver cancer sites, along with remodeling the tumor microenvironment, is crucial for enhancing the efficacy of chemotherapeutic agents. For this study, a platelet (PLT)-mediated nanodrug delivery system (DASA+ATO@PLT) was developed to improve the effectiveness of chemotherapy. This system delivers nano-sized dasatinib and atovaquone specifically to liver tumor sites and facilitates intra-tumoral permeation upon release. Through JC-1, immunohistochemistry, and DNA damage analyses, the therapeutic effect of DASA+ATO@PLT was assessed. In vitro simulation and intravital imaging were carried out to determine the accumulation of dasatinib and atovaquone in liver tumor sites. The experiment demonstrated the accumulation of dasatinib and atovaquone in tumor sites, followed by deep permeation in the tumor microenvironment with the assistance of PLTs, while simultaneously revealing the ability of DASA+ATO@PLT to remodel the liver cancer microenvironment (overcoming hypoxia) and enhance chemotherapeutic efficacy. This system utilizes the natural tumor recognition ability of PLTs and enhances the chemo-immunotherapeutic effect through targeted delivery of nano-chemotherapeutic drugs to the tumor, resulting in effective accumulation and infiltration. The PLT-mediated nanodrug delivery system serves as a “Trojan horse” to carry therapeutic drugs as cargo and deliver them to target cells, leading to favorable outcomes. Full article
Show Figures

Figure 1

21 pages, 7667 KiB  
Article
Single-Cell Profiling Reveals Immune-Based Mechanisms Underlying Tumor Radiosensitization by a Novel Mn Porphyrin Clinical Candidate, MnTnBuOE-2-PyP5+ (BMX-001)
by Sun Up Noh, Jinyeong Lim, Sung-Won Shin, Yeeun Kim, Woong-Yang Park, Ines Batinic-Haberle, Changhoon Choi and Won Park
Antioxidants 2024, 13(4), 477; https://doi.org/10.3390/antiox13040477 - 17 Apr 2024
Viewed by 2166
Abstract
Manganese porphyrins reportedly exhibit synergic effects when combined with irradiation. However, an in-depth understanding of intratumoral heterogeneity and immune pathways, as affected by Mn porphyrins, remains limited. Here, we explored the mechanisms underlying immunomodulation of a clinical candidate, MnTnBuOE-2-PyP5+ (BMX-001, MnBuOE), using [...] Read more.
Manganese porphyrins reportedly exhibit synergic effects when combined with irradiation. However, an in-depth understanding of intratumoral heterogeneity and immune pathways, as affected by Mn porphyrins, remains limited. Here, we explored the mechanisms underlying immunomodulation of a clinical candidate, MnTnBuOE-2-PyP5+ (BMX-001, MnBuOE), using single-cell analysis in a murine carcinoma model. Mice bearing 4T1 tumors were divided into four groups: control, MnBuOE, radiotherapy (RT), and combined MnBuOE and radiotherapy (MnBuOE/RT). In epithelial cells, the epithelial–mesenchymal transition, TNF-α signaling via NF-кB, angiogenesis, and hypoxia-related genes were significantly downregulated in the MnBuOE/RT group compared with the RT group. All subtypes of cancer-associated fibroblasts (CAFs) were clearly reduced in MnBuOE and MnBuOE/RT. Inhibitory receptor–ligand interactions, in which epithelial cells and CAFs interacted with CD8+ T cells, were significantly lower in the MnBuOE/RT group than in the RT group. Trajectory analysis showed that dendritic cells maturation-associated markers were increased in MnBuOE/RT. M1 macrophages were significantly increased in the MnBuOE/RT group compared with the RT group, whereas myeloid-derived suppressor cells were decreased. CellChat analysis showed that the number of cell–cell communications was the lowest in the MnBuOE/RT group. Our study is the first to provide evidence for the combined radiotherapy with a novel Mn porphyrin clinical candidate, BMX-001, from the perspective of each cell type within the tumor microenvironment. Full article
(This article belongs to the Section Natural and Synthetic Antioxidants)
Show Figures

Figure 1

15 pages, 4172 KiB  
Article
Onward Spread from Liver Metastases Is a Major Cause of Multi-Organ Metastasis in a Mouse Model of Metastatic Colon Cancer
by Liza A. Wijler, Bastiaan J. Viergever, Esther Strating, Susanne J. van Schelven, Susanna Poghosyan, Nicola C. Frenkel, Hedy te Rietmole, Andre Verheem, Danielle A. E. Raats, Inne H. M. Borel Rinkes, Jeroen Hagendoorn and Onno Kranenburg
Cancers 2024, 16(5), 1073; https://doi.org/10.3390/cancers16051073 - 6 Mar 2024
Cited by 6 | Viewed by 2991
Abstract
Colorectal cancer metastasizes predominantly to the liver but also to the lungs and the peritoneum. The presence of extra-hepatic metastases limits curative (surgical) treatment options and is associated with very poor survival. The mechanisms governing multi-organ metastasis formation are incompletely understood. Here, we [...] Read more.
Colorectal cancer metastasizes predominantly to the liver but also to the lungs and the peritoneum. The presence of extra-hepatic metastases limits curative (surgical) treatment options and is associated with very poor survival. The mechanisms governing multi-organ metastasis formation are incompletely understood. Here, we tested the hypothesis that the site of tumor growth influences extra-hepatic metastasis formation. To this end, we implanted murine colon cancer organoids into the primary tumor site (i.e., the caecum) and into the primary metastasis site (i.e., the liver) in immunocompetent mice. The organoid-initiated liver tumors were significantly more efficient in seeding distant metastases compared to tumors of the same origin growing in the caecum (intra-hepatic: 51 vs. 40%, p = 0.001; peritoneal cavity: 51% vs. 33%, p = 0.001; lungs: 30% vs. 7%, p = 0.017). The enhanced metastatic capacity of the liver tumors was associated with the formation of ‘hotspots’ of vitronectin-positive blood vessels surrounded by macrophages. RNA sequencing analysis of clinical samples showed a high expression of vitronectin in liver metastases, along with signatures reflecting hypoxia, angiogenesis, coagulation, and macrophages. We conclude that ‘onward spread’ from liver metastases is facilitated by liver-specific microenvironmental signals that cause the formation of macrophage-associated vascular hotspots. The therapeutic targeting of these signals may help to contain the disease within the liver and prevent onward spread. Full article
(This article belongs to the Topic Advances in Colorectal Cancer Therapy)
Show Figures

Figure 1

51 pages, 5674 KiB  
Review
Adjuvant Novel Nanocarrier-Based Targeted Therapy for Lung Cancer
by Kangkan Sarma, Md Habban Akther, Irfan Ahmad, Obaid Afzal, Abdulmalik S. A. Altamimi, Manal A. Alossaimi, Mariusz Jaremko, Abdul-Hamid Emwas and Preety Gautam
Molecules 2024, 29(5), 1076; https://doi.org/10.3390/molecules29051076 - 29 Feb 2024
Cited by 18 | Viewed by 5094
Abstract
Lung cancer has the lowest survival rate due to its late-stage diagnosis, poor prognosis, and intra-tumoral heterogeneity. These factors decrease the effectiveness of treatment. They release chemokines and cytokines from the tumor microenvironment (TME). To improve the effectiveness of treatment, researchers emphasize personalized [...] Read more.
Lung cancer has the lowest survival rate due to its late-stage diagnosis, poor prognosis, and intra-tumoral heterogeneity. These factors decrease the effectiveness of treatment. They release chemokines and cytokines from the tumor microenvironment (TME). To improve the effectiveness of treatment, researchers emphasize personalized adjuvant therapies along with conventional ones. Targeted chemotherapeutic drug delivery systems and specific pathway-blocking agents using nanocarriers are a few of them. This study explored the nanocarrier roles and strategies to improve the treatment profile’s effectiveness by striving for TME. A biofunctionalized nanocarrier stimulates biosystem interaction, cellular uptake, immune system escape, and vascular changes for penetration into the TME. Inorganic metal compounds scavenge reactive oxygen species (ROS) through their photothermal effect. Stroma, hypoxia, pH, and immunity-modulating agents conjugated or modified nanocarriers co-administered with pathway-blocking or condition-modulating agents can regulate extracellular matrix (ECM), Cancer-associated fibroblasts (CAF),Tyro3, Axl, and Mertk receptors (TAM) regulation, regulatory T-cell (Treg) inhibition, and myeloid-derived suppressor cells (MDSC) inhibition. Again, biomimetic conjugation or the surface modification of nanocarriers using ligands can enhance active targeting efficacy by bypassing the TME. A carrier system with biofunctionalized inorganic metal compounds and organic compound complex-loaded drugs is convenient for NSCLC-targeted therapy. Full article
Show Figures

Graphical abstract

13 pages, 5450 KiB  
Article
MicroRNA-204 Regulates Angiogenesis and Vasculogenic Mimicry in CD44+/CD24− Breast Cancer Stem-like Cells
by Martha Resendiz-Hernández, Alejandra P. García-Hernández, Macrina B. Silva-Cázares, Rogelio Coronado-Uribe, Olga N. Hernández-de la Cruz, Lourdes A. Arriaga-Pizano, Jessica L. Prieto-Chávez, Yarely M. Salinas-Vera, Eloisa Ibarra-Sierra, Concepción Ortiz-Martínez and César López-Camarillo
Non-Coding RNA 2024, 10(1), 14; https://doi.org/10.3390/ncrna10010014 - 9 Feb 2024
Cited by 9 | Viewed by 3515
Abstract
Tumors have high requirements in terms of nutrients and oxygen. Angiogenesis is the classical mechanism for vessel formation. Tumoral vascularization has the function of nourishing the cancer cells to support tumor growth. Vasculogenic mimicry, a novel intratumoral microcirculation system, alludes to the ability [...] Read more.
Tumors have high requirements in terms of nutrients and oxygen. Angiogenesis is the classical mechanism for vessel formation. Tumoral vascularization has the function of nourishing the cancer cells to support tumor growth. Vasculogenic mimicry, a novel intratumoral microcirculation system, alludes to the ability of cancer cells to organize in three-dimensional (3D) channel-like architectures. It also supplies the tumors with nutrients and oxygen. Both mechanisms operate in a coordinated way; however, their functions in breast cancer stem-like cells and their regulation by microRNAs remain elusive. In the present study, we investigated the functional role of microRNA-204 (miR-204) on angiogenesis and vasculogenic mimicry in breast cancer stem-like cells. Using flow cytometry assays, we found that 86.1% of MDA-MB-231 and 92% of Hs-578t breast cancer cells showed the CD44+/CD24− immunophenotype representative of cancer stem-like cells (CSCs). The MDA-MB-231 subpopulation of CSCs exhibited the ability to form mammospheres, as expected. Interestingly, we found that the restoration of miR-204 expression in CSCs significantly inhibited the number and size of the mammospheres. Moreover, we found that MDA-MB-231 and Hs-578t CSCs efficiently undergo angiogenesis and hypoxia-induced vasculogenic mimicry in vitro. The transfection of precursor miR-204 in both CSCs was able to impair the angiogenesis in the HUVEC cell model, which was observed as a diminution in the number of polygons and sprouting cells. Remarkably, miR-204 mimics also resulted in the inhibition of vasculogenic mimicry formation in MDA-MB-231 and Hs-578t CSCs, with a significant reduction in the number of channel-like structures and branch points. Mechanistically, the effects of miR-204 were associated with a diminution of pro-angiogenic VEGFA and β-catenin protein levels. In conclusion, our findings indicated that miR-204 abrogates the angiogenesis and vasculogenic mimicry development in breast cancer stem-like cells, suggesting that it could be a potential tool for breast cancer intervention based on microRNA replacement therapies. Full article
(This article belongs to the Section Detection and Biomarkers of Non-Coding RNA)
Show Figures

Figure 1

20 pages, 13405 KiB  
Article
MRI-Based Assessment of Brain Tumor Hypoxia: Correlation with Histology
by Fatemeh Arzanforoosh, Maaike Van der Velden, Avery J. L. Berman, Sebastian R. Van der Voort, Eelke M. Bos, Joost W. Schouten, Arnaud J. P. E. Vincent, Johan M. Kros, Marion Smits and Esther A. H. Warnert
Cancers 2024, 16(1), 138; https://doi.org/10.3390/cancers16010138 - 27 Dec 2023
Cited by 9 | Viewed by 2362
Abstract
Cerebral hypoxia significantly impacts the progression of brain tumors and their resistance to radiotherapy. This study employed streamlined quantitative blood-oxygen-level-dependent (sqBOLD) MRI to assess the oxygen extraction fraction (OEF)—a measure of how much oxygen is being extracted from vessels, with higher OEF values [...] Read more.
Cerebral hypoxia significantly impacts the progression of brain tumors and their resistance to radiotherapy. This study employed streamlined quantitative blood-oxygen-level-dependent (sqBOLD) MRI to assess the oxygen extraction fraction (OEF)—a measure of how much oxygen is being extracted from vessels, with higher OEF values indicating hypoxia. Simultaneously, we utilized vessel size imaging (VSI) to evaluate microvascular dimensions and blood volume. A cohort of ten patients, divided between those with glioma and those with brain metastases, underwent a 3 Tesla MRI scan. We generated OEF, cerebral blood volume (CBV), and vessel size maps, which guided 3–4 targeted biopsies per patient. Subsequent histological analyses of these biopsies used hypoxia-inducible factor 1-alpha (HIF-1α) for hypoxia and CD31 for microvasculature assessment, followed by a correlation analysis between MRI and histological data. The results showed that while the sqBOLD model was generally applicable to brain tumors, it demonstrated discrepancies in some metastatic tumors, highlighting the need for model adjustments in these cases. The OEF, CBV, and vessel size maps provided insights into the tumor’s hypoxic condition, showing intertumoral and intratumoral heterogeneity. A significant relationship between MRI-derived measurements and histological data was only evident in the vessel size measurements (r = 0.68, p < 0.001). Full article
(This article belongs to the Special Issue Advances in Neuro-Oncological Imaging)
Show Figures

Figure 1

Back to TopTop