Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (188)

Search Parameters:
Keywords = in vivo breast imaging

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
10 pages, 1468 KiB  
Article
Noninvasive Mapping of Extracellular Potassium in Breast Tumors via Multi-Wavelength Photoacoustic Imaging
by Jeff Folz, Ahmad Eido, Maria E. Gonzalez, Roberta Caruso, Xueding Wang, Celina G. Kleer and Janggun Jo
Sensors 2025, 25(15), 4724; https://doi.org/10.3390/s25154724 - 31 Jul 2025
Viewed by 226
Abstract
Elevated extracellular potassium (K+) in the tumor microenvironment (TME) of breast and other cancers is increasingly recognized as a critical factor influencing tumor progression and immune suppression. Current methods for noninvasive mapping of the potassium distribution in tumors are limited. Here, [...] Read more.
Elevated extracellular potassium (K+) in the tumor microenvironment (TME) of breast and other cancers is increasingly recognized as a critical factor influencing tumor progression and immune suppression. Current methods for noninvasive mapping of the potassium distribution in tumors are limited. Here, we employed photoacoustic chemical imaging (PACI) with a solvatochromic dye-based, potassium-sensitive nanoprobe (SDKNP) to quantitatively visualize extracellular potassium levels in an orthotopic metaplastic breast cancer mouse model, Ccn6-KO. Tumors of three distinct sizes (5 mm, 10 mm, and 20 mm) were imaged using multi-wavelength photoacoustic imaging at five laser wavelengths (560, 576, 584, 605, and 625 nm). Potassium concentration maps derived from spectral unmixing of the photoacoustic images at the five laser wavelengths revealed significantly increased potassium levels in larger tumors, confirmed independently by inductively coupled plasma mass spectrometry (ICP-MS). The PACI results matched ICP-MS measurements, validating PACI as a robust, noninvasive imaging modality for potassium mapping in tumors in vivo. This work establishes PACI as a promising tool for studying the chemical properties of the TME and provides a foundation for future studies evaluating the immunotherapy response through ionic biomarker imaging. Full article
(This article belongs to the Special Issue Advances in Photoacoustic Resonators and Sensors)
Show Figures

Figure 1

16 pages, 5172 KiB  
Article
LAMP1 as a Target for PET Imaging in Adenocarcinoma Xenograft Models
by Bahar Ataeinia, Arvin Haj-Mirzaian, Lital Ben-Naim, Shadi A. Esfahani, Asier Marcos Vidal, Umar Mahmood and Pedram Heidari
Pharmaceuticals 2025, 18(8), 1122; https://doi.org/10.3390/ph18081122 - 27 Jul 2025
Viewed by 512
Abstract
Background: Lysosomal-associated membrane protein 1 (LAMP1), typically localized to the lysosomal membrane, is increasingly implicated as a marker of cancer aggressiveness and metastasis when expressed on the cell surface. This study aimed to develop a LAMP1-targeted antibody-based PET tracer and assess its efficacy [...] Read more.
Background: Lysosomal-associated membrane protein 1 (LAMP1), typically localized to the lysosomal membrane, is increasingly implicated as a marker of cancer aggressiveness and metastasis when expressed on the cell surface. This study aimed to develop a LAMP1-targeted antibody-based PET tracer and assess its efficacy in mouse models of human breast and colon adenocarcinoma. Methods: To determine the source of LAMP1 expression, we utilized human single-cell RNA sequencing and spatial transcriptomics, complemented by in-house flow cytometry on xenografted mouse models. Tissue microarrays of multiple epithelial cancers and normal tissue were stained for LAMP-1, and staining was quantified. An anti-LAMP1 monoclonal antibody was conjugated with desferrioxamine (DFO) and labeled with zirconium-89 (89Zr). Human triple-negative breast cancer (MDA-MB-231) and colon cancer (Caco-2) cell lines were implanted in nude mice. PET/CT imaging was conducted at 24, 72, and 168 h post-intravenous injection of 89Zr-DFO-anti-LAMP1 and 89Zr-DFO-IgG (negative control), followed by organ-specific biodistribution analyses at the final imaging time point. Results: Integrated single-cell and spatial RNA sequencing demonstrated that LAMP1 expression was localized to myeloid-derived suppressor cells (MDSCs) and cancer-associated fibroblasts (CAFs) in addition to the cancer cells. Tissue microarray showed significantly higher staining for LAMP-1 in tumor tissue compared to normal tissue (3986 ± 2635 vs. 1299 ± 1291, p < 0.001). Additionally, xenograft models showed a significantly higher contribution of cancer cells than the immune cells to cell surface LAMP1 expression. In vivo, PET imaging with 89Zr-DFO-anti-LAMP1 PET/CT revealed detectable tumor uptake as early as 24 h post-injection. The 89Zr-DFO-anti-LAMP1 tracer demonstrated significantly higher uptake than the control 89Zr-DFO-IgG in both models across all time points (MDA-MB-231 SUVmax at 168 h: 12.9 ± 5.7 vs. 4.4 ± 2.4, p = 0.003; Caco-2 SUVmax at 168 h: 8.53 ± 3.03 vs. 3.38 ± 1.25, p < 0.01). Conclusions: Imaging of cell surface LAMP-1 in breast and colon adenocarcinoma is feasible by immuno-PET. LAMP-1 imaging can be expanded to adenocarcinomas of other origins, such as prostate and pancreas. Full article
Show Figures

Figure 1

19 pages, 3398 KiB  
Article
Synthesis and Evaluation of [18F]AlF-NOTA-iPD-L1 as a Potential Theranostic Pair for [177Lu]Lu-DOTA-iPD-L1
by Guillermina Ferro-Flores, Myrna Luna-Gutiérrez, Blanca Ocampo-García, Nallely Jiménez-Mancilla, Nancy Lara-Almazán, Rigoberto Oros-Pantoja, Clara Santos-Cuevas, Erika Azorín-Vega and Laura Meléndez-Alafort
Pharmaceutics 2025, 17(7), 920; https://doi.org/10.3390/pharmaceutics17070920 - 16 Jul 2025
Viewed by 399
Abstract
Background/Objective: Programmed cell death ligand-1 (PD-L1), which is overexpressed in certain tumors, inhibits the body’s natural immune response by providing an “off” signal that enables cancer cells to evade the immune system. It has been demonstrated that [177Lu]Lu-DOTA-iPD-L1 (PD-L1 inhibitor [...] Read more.
Background/Objective: Programmed cell death ligand-1 (PD-L1), which is overexpressed in certain tumors, inhibits the body’s natural immune response by providing an “off” signal that enables cancer cells to evade the immune system. It has been demonstrated that [177Lu]Lu-DOTA-iPD-L1 (PD-L1 inhibitor cyclic peptide) promotes immune responses. This study aimed to synthesize and evaluate [18F]AlF-NOTA-iPD-L1 as a novel radiotracer for PD-L1 positron emission tomography (PET) imaging and as a potential theranostic pair for [177Lu]Lu-DOTA-iPD-L1. Methods: The NOTA-iPD-L1 peptide conjugate was synthesized and characterized by U.V.-vis, I.R.-FT, and UPLC-mass spectroscopies. Radiolabeling was performed using [18F]AlF as the precursor, and the radiochemical purity (HPLC), partition coefficient, and serum stability were assessed. Cellular uptake and internalization (in 4T1 triple-negative breast cancer cells), binding competition, immunofluorescence, and Western blot assays were applied for the radiotracer in vitro characterization. Biodistribution in mice bearing 4T1 tumors was performed, and molecular imaging (Cerenkov images) of [18F]AlF-NOTA-iPD-L1 and [177Lu]Lu-DOTA-iPD-L1 in the same mouse was obtained. Results: [18F]AlF-NOTA-iPD-L1 was prepared with a radiochemical purity greater than 97%, and it demonstrated high in vitro and in vivo stability, as well as specific recognition by the PD-L1 protein (IC50 = 9.27 ± 2.69 nM). Biodistribution studies indicated a tumor uptake of 6.4% ± 0.9% ID/g at 1-hour post-administration, and Cerenkov images showed a high tumor uptake of both [18F]AlF-NOTA-iPD-L1 and 177Lu-iPD-L1 in the same mouse. Conclusions: These results warrant further studies to evaluate the clinical usefulness of [18F]AlF-NOTA-iPD-L1/[177Lu]Lu-DOTA-iPD-L1 as a radiotheranostic pair in combination with anti-PD-L1/PD1 immunotherapy. Full article
Show Figures

Figure 1

20 pages, 4435 KiB  
Article
89Zr-Radiolabelling of p-NCS-Bz-DFO-Anti-HER2 Affibody Immunoconjugate: Characterization and Assessment of In Vitro Potential in HER2-Positive Breast Cancer Imaging
by Maria-Roxana Tudoroiu-Cornoiu, Radu Marian Șerban, Diana Cocioabă, Dragoș Andrei Niculae, Doina Drăgănescu, Radu Leonte, Alina Catrinel Ion and Dana Niculae
Pharmaceutics 2025, 17(6), 739; https://doi.org/10.3390/pharmaceutics17060739 - 4 Jun 2025
Viewed by 671
Abstract
Background: The 89Zr radioisotope is increasingly vital in positron emission tomography (PET), especially immuno-PET, due to its long half-life of 78.4 h, allowing extended tracking of biological processes. This makes it particularly suitable for researching medicines with slow pharmacokinetics and enhances the [...] Read more.
Background: The 89Zr radioisotope is increasingly vital in positron emission tomography (PET), especially immuno-PET, due to its long half-life of 78.4 h, allowing extended tracking of biological processes. This makes it particularly suitable for researching medicines with slow pharmacokinetics and enhances the precision of molecular imaging, especially in oncology. Despite zirconium’s potential for skeletal accumulation, effective chelation with agents like deferoxamine (DFO) enables high-resolution imaging of antigen-specific tumours, such as HER2-positive breast cancer, offering insights into tumour biology and treatment response. Methods: 89Zr was produced at the ACSI TR-19 cyclotron via 89Y(p,n)89Zr reaction. Natural yttrium foils (250 μm) were irradiated with 12.9 MeV protons on target, with 100 μA·h. An HER2-targeting affibody was synthesized and conjugated with p-NCS-Bz-DFO (1:4 mass ratio) at 37 °C for 60 min (pH 9.2 ± 0.2), then purified on a PD-10 column. Radiolabelling was performed with [89Zr]Zr-oxalate at pH ranging from 7.0 to 9.0, with concentrations from 110 to 460 MBq/mL. Results: Final activity reached 2.95 ± 0.31 GBq/batch (EOB corrected), with ≥ 99.9% radionuclide and ≥95% radiochemical purities. The anti-HER2 affibody was successfully radiolabelled with 89Zr, resulting in a radiochemical purity of over 85% with molar activity of 26.5 ± 4.4 and 11.45 MBq/nmol at pH 7.0–7.5. In vitro tests on BT-474 and MCF-7 cell lines confirmed high uptake in HER2-positive cells, validating specificity and stability. Conclusions: The successful synthesis and labelling of the [89Zr]Zr-p-NCS-Bz-DFO-anti-HER2 affibody are promising achievements for its further application in targeted immuno-PET imaging for HER2-positive malignancies. Further in vivo studies are needed to support its clinical translation. Full article
(This article belongs to the Section Physical Pharmacy and Formulation)
Show Figures

Figure 1

16 pages, 3209 KiB  
Article
Side-Illuminating Optical Fiber for High-Power-Density-Mediated Intraluminal Photoacoustic Imaging
by Nidhi Singh, Carlos-Felipe Roa, Emmanuel Chérin, Lothar Lilge and Christine E. M. Demore
Appl. Sci. 2025, 15(7), 3639; https://doi.org/10.3390/app15073639 - 26 Mar 2025
Viewed by 1063
Abstract
Intraluminal photoacoustic (PA) imaging has the potential for providing physiological and functional information in wide-ranging clinical applications. Along with endoluminal ultrasound transducers, these applications require compact light delivery devices which can deliver high-energy ns-pulsed laser to the target region. In this work, we [...] Read more.
Intraluminal photoacoustic (PA) imaging has the potential for providing physiological and functional information in wide-ranging clinical applications. Along with endoluminal ultrasound transducers, these applications require compact light delivery devices which can deliver high-energy ns-pulsed laser to the target region. In this work, we describe the design, method of fabrication and characterization of a new compact, side-fire optical fiber that can deliver high-energy laser pulses for PA imaging. Side-fire illuminators were fabricated using UV laser ablation to create windows on the side of a 1.5 mm diameter single core, multi-mode optical fiber with a reflective silver coating and a beveled end. Devices with 10 mm, 20 mm, and 30 mm window lengths were fabricated and their beam profiles characterized. Elongated side-fire fibers with −6 dB beam size up to 30.79 mm × 5.5 mm were developed. A side-fire to total output ratio of up to 0.69 and a side fire efficiency of up to 40%, relative to a standard front-fire fiber, were achieved. We evaluated the effects of high-energy ns-pulsed light propagation on the fiber by coupling the fiber to 18 mJ or 100 MW/cm2 (at 750 nm) beam from a Q-switched laser. The PA imaging with the fiber was demonstrated by detecting India ink targets embedded in chicken breast tissue over the full length of a 20 mm illumination window and over a 100° angle and by visualizing in vivo the rat ear vasculature. Full article
(This article belongs to the Special Issue Advanced Optical-Fiber-Related Technologies)
Show Figures

Figure 1

16 pages, 3216 KiB  
Article
Multifaceted Functional Liposomes: Theranostic Potential of Liposomal Indocyanine Green and Doxorubicin for Enhanced Anticancer Efficacy and Imaging
by Wei-Ting Liao, Dao-Ming Chang, Meng-Xian Lin, Te-Sen Chou, Yi-Chung Tung and Jong-Kai Hsiao
Pharmaceutics 2025, 17(3), 344; https://doi.org/10.3390/pharmaceutics17030344 - 7 Mar 2025
Viewed by 1004
Abstract
Background/Objectives: Liposomal drug formulations improve anticancer treatment efficacy and reduce toxicity by altering pharmacokinetics and biodistribution. Indocyanine Green (ICG), an FDA-approved near-infrared imaging agent, exhibits photosensitivity, photothermal effects, and potential ferroptosis induction, enhancing anticancer activity. Doxorubicin (DOX), widely used for treating breast, ovarian, [...] Read more.
Background/Objectives: Liposomal drug formulations improve anticancer treatment efficacy and reduce toxicity by altering pharmacokinetics and biodistribution. Indocyanine Green (ICG), an FDA-approved near-infrared imaging agent, exhibits photosensitivity, photothermal effects, and potential ferroptosis induction, enhancing anticancer activity. Doxorubicin (DOX), widely used for treating breast, ovarian, and liver cancers, is limited by cardiotoxicity, requiring dosage control. Incorporating ICG and DOX into liposomes enables medical imaging, controlled drug release, reduced administration frequency, and fewer side effects. This study aims to develop liposomes encapsulating both ICG and DOX and evaluate their theranostic potential in in vitro and in vivo lung adenocarcinoma models. Methods: Liposomes containing ICG and DOX (Lipo-ICG/DOX) were synthesized using an active loading method and characterized for size (~140 nm), lipid, and drug concentrations. In vitro studies using A549 lung cancer cells assessed liposome uptake via fluorescence microscopy, while in vivo xenograft models evaluated therapeutic efficacy. Results: Lipo-ICG/DOX showed uptake in A549 cells, with ICG localizing in lysosomes and DOX in nuclei. Treatment reduced cell viability significantly by day three. In vivo imaging demonstrated the retention of liposomes in tumor sites, with ICG signals observed in the liver and intestines, indicating metabolic routes. When combined with 780 nm light exposure, liposomes slowed tumor growth over 12 days. Mechanistic studies revealed combined ferroptosis and apoptosis induction. Conclusions: Lipo-ICG/DOX demonstrates strong theranostic potential, integrating imaging and therapy for lung adenocarcinoma. This multifunctional formulation offers a promising strategy for improving cancer treatment efficacy while minimizing side effects. Full article
Show Figures

Graphical abstract

20 pages, 30831 KiB  
Article
Monensin Inhibits Triple-Negative Breast Cancer in Mice by a Na+-Dependent Cytotoxic Action Unrelated to Cytostatic Effects
by Simone Fiorilla, Francesco Tasso, Nausicaa Clemente, Tommaso Trisciuoglio, Renzo Boldorini and Rita Carini
Cells 2025, 14(3), 185; https://doi.org/10.3390/cells14030185 - 25 Jan 2025
Cited by 2 | Viewed by 1342
Abstract
Triple-negative breast cancer (TNBC) represents the most aggressive breast cancer subtype, defined by its limited therapeutic options and poor outcomes. This study investigated the therapeutic potential of targeting Na+ homeostasis in TNBC cells to induce TNBC inhibition. For this purpose, BALB/c mice [...] Read more.
Triple-negative breast cancer (TNBC) represents the most aggressive breast cancer subtype, defined by its limited therapeutic options and poor outcomes. This study investigated the therapeutic potential of targeting Na+ homeostasis in TNBC cells to induce TNBC inhibition. For this purpose, BALB/c mice were inoculated with 4T1-Luc2 breast cancer cells and treated with the Na+ ionophore monensin (8 mg/kg) or vehicle alone. Tumor development and cellular Na+ content were assessed using vivo live imaging techniques, while intracellular Na+ variations and cytotoxicity were evaluated through live cell analysis. Monensin treatment increased Na+ levels in cancerous tissues and reduced TNBC mass (monensin: 0.146 ± 0.06; vehicle: 0.468 ± 0.2 cm3; p < 0.001). This treatment induced extensive necrosis in TNBC tumors while preserving the structural and functional integrity of healthy organs and maintaining the proliferative activity of both tumor and normal tissues. Monensin did not alter the expression of proliferating nuclear antigen (PCNA) in 4T1-Luc2 cells but triggered cytotoxicity preceded by intracellular Na+ accumulation. Na+-free conditions prevented both Na+ accumulation and 4T1-Luc2 cell death. Thus, monensin exerts its antitumor effects in TNBC through a Na+-dependent and tumor-specific cytotoxic mechanism, without inducing cytostatic effects on normal or transformed tissues. Collectively, these findings underscore the potential of Na+ ionophores as promising therapeutic agents for TNBC. Full article
(This article belongs to the Section Cellular Pathology)
Show Figures

Figure 1

17 pages, 3743 KiB  
Article
Pegylated NIR Fluorophore-Conjugated OBHSA Prodrug for ERα-Targeted Theranostics with Enhanced Imaging and Long-Term Retention
by Xiaohua Wang, Xiaofei Deng, Lilan Xin, Chune Dong, Guoyuan Hu and Hai-Bing Zhou
Molecules 2025, 30(2), 305; https://doi.org/10.3390/molecules30020305 - 14 Jan 2025
Viewed by 1062
Abstract
In recent years, the near-infrared (NIR) fluorescence theranostic system has garnered increasing attention for its advantages in the simultaneous diagnosis- and imaging-guided delivery of therapeutic drugs. However, challenges such as strong background fluorescence signals and rapid metabolism have hindered the achievement of sufficient [...] Read more.
In recent years, the near-infrared (NIR) fluorescence theranostic system has garnered increasing attention for its advantages in the simultaneous diagnosis- and imaging-guided delivery of therapeutic drugs. However, challenges such as strong background fluorescence signals and rapid metabolism have hindered the achievement of sufficient contrast between tumors and surrounding tissues, limiting the system’s applicability. This study aims to integrate the pegylation strategy with a tumor microenvironment-responsive approach. A novel esterase-activated EPR strategy prodrug, OBHSA-PEG-DCM, was designed. This prodrug links OBHSA, a protein degrader capable of efficient ERα protein degradation, to the PEG-modified fluorescent group (dicyanomethylene-4H-pyran, DCM) via an ester bond. This integration facilitates targeted drug delivery and enhances the retention of the fluorescent group within the tumor, allowing distinct in vivo tumor imaging periods. Experimental results show that, benefiting from overexpressed esterase in cancer cells, OBHSA-PEG-DCM can be efficiently hydrolyzed, releasing OBHSA and pegylated DCM. OBHSA demonstrated potent inhibition against MCF-7 cells (IC50 = 1.09 μM). Simultaneously, pegylated DCM exhibited remarkable in vivo imaging capabilities, lasting up to 12 days in mice, due to the enhanced permeability and retention (EPR) effect. OBHSA-PEG-DCM holds promise as a theranostic agent for ERα-positive breast cancer, offering both therapeutic and diagnostic capabilities. Importantly, this study highlights the utility of pegylated NIR fluorophores for long-circulating drug delivery systems, addressing current challenges in achieving high-contrast tumor imaging and effective targeted drug release. Full article
Show Figures

Figure 1

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 - 21 Dec 2024
Cited by 4 | Viewed by 1696
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

14 pages, 3727 KiB  
Article
Engineering Zn/Fe Mixed Metal Oxides with Tunable Structural and Magnetic Properties for Magnetic Particle Imaging
by Qianyi Zhang, Bing Sun, Saeed Shanehsazzadeh, Andre Bongers and Zi Gu
Nanomaterials 2024, 14(23), 1964; https://doi.org/10.3390/nano14231964 - 7 Dec 2024
Cited by 2 | Viewed by 1352
Abstract
Engineering magnetic nanoparticles with tunable structural properties and magnetism is critical to develop desirable magnetic particle imaging (MPI) tracers for biomedical applications. Here we present a new superparamagnetic metal oxide nanoparticle with a controllable chemical composition and magnetism for imaging tumor xenografts in [...] Read more.
Engineering magnetic nanoparticles with tunable structural properties and magnetism is critical to develop desirable magnetic particle imaging (MPI) tracers for biomedical applications. Here we present a new superparamagnetic metal oxide nanoparticle with a controllable chemical composition and magnetism for imaging tumor xenografts in living mice. Superparamagnetic Zn/Fe mixed metal oxide (ZnFe-MMO) nanoparticles are fabricated via a facile one-pot co-precipitation method in water followed by thermal decomposition with tunable Zn/Fe ratios and at various calcination temperatures. This work, for the first time, presented LDH-derived metal oxides for an MPI application. The metal composition is tunable to present an optimized MPI performance. The analytical results demonstrate that ZnFe-MMO nanoparticles at the designed molar ratio of Zn/Fe = 2:1 after 650 °C calcination demonstrate a higher saturation magnetization (MS) value and optimal MPI signal than the samples presented with other conditions. The excellent biocompatibility of ZnFe-MMO is demonstrated in both breast cancer cells and fibroblast cell cultures. In vivo imaging of 4T1 tumor xenografts in mice using ZnFe-MMO as a tracer showed that the mean signal intensity is 1.27-fold higher than the commercial tracer VivoTrax at 72 h post-injection, indicating ZnFe-MMO’s promise for prolonged MPI imaging applications. Full article
Show Figures

Figure 1

16 pages, 3709 KiB  
Article
Polymeric Nanoparticles Potentiate the Anticancer Activity of Novel PI3Kα Inhibitors Against Triple-Negative Breast Cancer Cells
by Suhair Sunoqrot, Samah Abusulieh and Dima Sabbah
Biomedicines 2024, 12(12), 2676; https://doi.org/10.3390/biomedicines12122676 - 24 Nov 2024
Cited by 1 | Viewed by 1172
Abstract
Background: Dysregulation in phosphoinositide-3-kinase alpha (PI3Kα) signaling is implicated in the development of various cancers, including triple-negative breast cancer (TNBC). We have previously synthesized a series of N-phenyl-6-chloro-4-hydroxy-2-quinolone-3-carboxamides as targeted inhibitors against PI3Kα. Herein, two drug candidates, R7 and R11, were selected [...] Read more.
Background: Dysregulation in phosphoinositide-3-kinase alpha (PI3Kα) signaling is implicated in the development of various cancers, including triple-negative breast cancer (TNBC). We have previously synthesized a series of N-phenyl-6-chloro-4-hydroxy-2-quinolone-3-carboxamides as targeted inhibitors against PI3Kα. Herein, two drug candidates, R7 and R11, were selected to be further investigated as a nanoparticle (NP) formulation against TNBC. Methods: R7 and R11 were entrapped in D-α-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS) polymeric NPs by nanoprecipitation. Following their physicochemical characterization, the anticancer activity of the compounds and their NP formulations was evaluated in the TNBC cell line MDA-MB-231 by conducting viability, uptake, and apoptosis assays, as well as penetration assays in a multicellular tumor spheroid model. Results: The NPs exhibited a particle size of 100–200 nm, excellent drug loading efficiencies, and sustained release under physiologic conditions. Viability assays revealed superior potency for the NP formulations, with IC50 values of 20 µM and 30 µM for R7- and R11-loaded NPs, respectively, compared to the free compounds, which exhibited IC50 values of 280 µM and 290 µM for R7 and R11, respectively. These results were attributed to the inherent antiproliferative activity of TPGS, as evidenced by the cytotoxicity of the drug-free NPs, as well as the enhanced cellular uptake enabled by the NP vehicle, as demonstrated by fluorescence microscopy imaging and flow cytometry measurements. Further investigations showed that the NPs promoted apoptosis via a mitochondrial-dependent pathway that involved the activation of proapoptotic caspases. Moreover, the NP formulations enhanced the penetration ability of the free compounds in multicellular tumor spheroids, causing a time- and concentration-dependent disruption of the spheroids. Conclusions: Our findings highlight the important role nanotechnology can play in improving the biopharmaceutical properties of new drug candidates and facilitating their in vivo translation. Full article
(This article belongs to the Special Issue Drug Resistance and Novel Targets for Cancer Therapy—Second Edition)
Show Figures

Graphical abstract

20 pages, 5713 KiB  
Article
A Comparison of the Sensitivity and Cellular Detection Capabilities of Magnetic Particle Imaging and Bioluminescence Imaging
by Sophia Trozzo, Bijita Neupane and Paula J. Foster
Tomography 2024, 10(11), 1846-1866; https://doi.org/10.3390/tomography10110135 - 20 Nov 2024
Cited by 2 | Viewed by 2136
Abstract
Background: Preclinical cell tracking is enhanced with a multimodal imaging approach. Bioluminescence imaging (BLI) is a highly sensitive optical modality that relies on engineering cells to constitutively express a luciferase gene. Magnetic particle imaging (MPI) is a newer imaging modality that directly detects [...] Read more.
Background: Preclinical cell tracking is enhanced with a multimodal imaging approach. Bioluminescence imaging (BLI) is a highly sensitive optical modality that relies on engineering cells to constitutively express a luciferase gene. Magnetic particle imaging (MPI) is a newer imaging modality that directly detects superparamagnetic iron oxide (SPIO) particles used to label cells. Here, we compare BLI and MPI for imaging cells in vitro and in vivo. Methods: Mouse 4T1 breast carcinoma cells were transduced to express firefly luciferase, labeled with SPIO (ProMag), and imaged as cell samples after subcutaneous injection into mice. Results: For cell samples, the BLI and MPI signals were strongly correlated with cell number. Both modalities presented limitations for imaging cells in vivo. For BLI, weak signal penetration, signal attenuation, and scattering prevented the detection of cells for mice with hair and for cells far from the tissue surface. For MPI, background signals obscured the detection of low cell numbers due to the limited dynamic range, and cell numbers could not be accurately quantified from in vivo images. Conclusions: It is important to understand the shortcomings of these imaging modalities to develop strategies to improve cellular detection sensitivity. Full article
Show Figures

Figure 1

17 pages, 2881 KiB  
Article
Immediate Diagnosis of Breast Carcinoma on Core Needle Biopsy Using Ex Vivo Fluorescence Confocal Microscopy: Feasibility in a One-Stop Breast Clinic Workflow
by Marie-Christine Mathieu, Voichita Suciu, Marie-Laure Tanguy, Neila Ines Ben Romdhane, Salma Moalla, Sana Harguem-Zayani, Remy Barbe, Corinne Balleyguier, Angelica Conversano and Muriel Abbaci
Life 2024, 14(11), 1384; https://doi.org/10.3390/life14111384 - 28 Oct 2024
Cited by 1 | Viewed by 1443
Abstract
Background: In the one-stop breast clinic setting, breast cytology traditionally provides immediate diagnosis of carcinoma. Fluorescence confocal microscopy (FCM) is an emerging optical technique enabling ex vivo analysis of breast biopsies in real-time. This study represents the first proof of concept for integrating [...] Read more.
Background: In the one-stop breast clinic setting, breast cytology traditionally provides immediate diagnosis of carcinoma. Fluorescence confocal microscopy (FCM) is an emerging optical technique enabling ex vivo analysis of breast biopsies in real-time. This study represents the first proof of concept for integrating FCM imaging into the routine workflow of breast core needle biopsies (CNB) at Gustave Roussy’s one-stop breast clinic. Methods: Fifty women with breast masses underwent consecutive enrollment. Biopsies were stained with acridine orange and fast green, followed by imaging using the Vivascope 2500M-G4 (FCM). Interpretation was conducted by two pathologists in real time (PT1) or postoperatively (PT2). Concordance with definitive histology, the duration of the FCM protocol, and its impact on conventional histopathology, immunohistochemistry, and FISH analyses were evaluated. Results: In our study of 50 biopsies, a concordant diagnosis of malignancy was performed using FCM on the malignant cases at definitive histology in 93.5% (29/31 cases) and in 90.3% (28/31 cases) according to PT1 and PT2, respectively. When the FCM suspicious cases were added, FCM identified 100% (31/31 cases) and 96.7% (30/31 cases) of the malignant cases according to PT1 and PT2, respectively. A notable false positive case was identified as a complex sclerosing lesion. The median time for sample preparation (including tissue reception) was 5 min, while the median time for imaging acquisition with interpretation was 3 min for PT1, but 1 min required for interpretation alone by PT2. Histopathological alterations were not more prevalent in FCM-imaged biopsies compared to conventionally treated biopsies. The immunophenotyping and molecular assessment of tissue were preserved after FCM protocol. Conclusions: FCM shows promise as a new histological method for the immediate diagnosis of breast carcinoma on core needle biopsies in a one-stop clinic setting, while also preserving tissue specimens for final histology. Full article
Show Figures

Figure 1

14 pages, 422 KiB  
Review
Micro-CT Microcalcification Analysis: A Scoping Review of Current Applications and Future Potential in Breast Cancer Research
by Redona Brahimetaj, Jan Cornelis and Bart Jansen
Tomography 2024, 10(11), 1716-1729; https://doi.org/10.3390/tomography10110126 - 24 Oct 2024
Cited by 2 | Viewed by 2281
Abstract
Micro-computed tomography (micro-CT) is a non-destructive imaging technique that offers highly detailed, 3D visualizations of a target specimen. In the context of breast cancer, micro-CT has emerged as a promising tool for analyzing microcalcifications (MCs), tiny calcium deposits that can indicate at an [...] Read more.
Micro-computed tomography (micro-CT) is a non-destructive imaging technique that offers highly detailed, 3D visualizations of a target specimen. In the context of breast cancer, micro-CT has emerged as a promising tool for analyzing microcalcifications (MCs), tiny calcium deposits that can indicate at an early stage the presence of cancer. This review aimed to explore the current applications of micro-CT in analyzing breast MCs (ex vivo, animal models, and phantoms) and to identify potential avenues in scientific research. We followed PRISMA guidelines for scoping reviews, yielding 18 studies that met our criteria. The studies varied in their purposes: feasibility and optimization of micro-CT for breast cancer imaging and MC analysis/diagnosis, comparison with other imaging modalities, development of micro-CT scanners and processing algorithms, enhancement of MC detection through contrast agents, etc. In conclusion, micro-CT offers superior image quality and detailed visualization of breast tissue (especially tumor masses and MCs), surpassing traditional methods like mammography and approaching the level of detail of histology. It holds great potential to enhance our understanding of MC characteristics and breast pathologies when used as a supplementary tool. Further research will solidify its role in clinical practice and potentially expand its applications in breast cancer studies. Full article
Show Figures

Figure 1

17 pages, 7781 KiB  
Article
Lymphovascular Tumoral Emboli in Inflammatory Breast Cancer Result from Haptotaxis-Mediated Encircling Lymphangiogenesis
by Justin Wang, Robert M. Hoffman, Yin Ye, Jordan Dillard and Sanford H. Barsky
Lymphatics 2024, 2(4), 195-211; https://doi.org/10.3390/lymphatics2040016 - 8 Oct 2024
Cited by 1 | Viewed by 2655
Abstract
Inflammatory breast cancer (IBC) is characterized by numerous tumor emboli within lymphatics. In a recent study, we observed tumor embolic budding both in vitro and in vivo within lymphovascular spaces and proposed this to account for the plethora of tumor emboli seen in [...] Read more.
Inflammatory breast cancer (IBC) is characterized by numerous tumor emboli within lymphatics. In a recent study, we observed tumor embolic budding both in vitro and in vivo within lymphovascular spaces and proposed this to account for the plethora of tumor emboli seen in IBC. These observations did not address, however, how lymphovascular invasion is initiated or the mechanisms involved. In the present study, using the well-characterized patient-derived xenograft (PDX), Mary-X, which exhibited florid lymphovascular invasion (LVI) in athymic mice (LVI) as defined by E-cadherin-positive tumor emboli within lymphatic channels distinguished by podoplanin and LYVE1 membrane and Prox1 nuclear immunoreactivities and spontaneous spheroidgenesis in vitro and human cases of IBC which showed similar LVI, we compared laser-captured microdissected emboli from Mary-X and from the cases of human IBC to non-embolic areas. Mary-X and IBC emboli expressed high levels of E-cadherin and no evidence of epithelial–mesenchymal transition (EMT). Mary-X spheroids expressed high levels of VEGF, especially VEGF-C, and stimulated both vascular and lymphatic endothelial haptotaxis. We then transplanted Mary-X serially into green, cyano, red, and nestin-green fluorescing protein (GFP-, CFP-, RFP-, and nestin-GFP) transgenic reporter mice in various combinations. Multicolor murine imaging studies indicated that reporter-labeled stroma initially encircled clumps of tumor cells and then served as a scaffold that supported nestin-GFP-labeled endothelial haptotaxis resulting in encircling lymphangiogenesis, confirmed by dual LYVE1 immunofluorescence. The present studies demonstrate a possible mechanism of a critical step of the tumor emboli formation of IBC. Full article
Show Figures

Figure 1

Back to TopTop