Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (860)

Search Parameters:
Keywords = cancer theranostic

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 808 KB  
Review
Albumin Nanoparticles as Multifunctional Carriers for Advanced Therapeutics
by Bogusława Konopska, Janusz Sokołowski, Anna Woźniak, Mikołaj Kondracki, Jakub Federowicz, Wojciech Grodzki, Agnieszka Bronowicka-Szydełko and Katarzyna Madziarska
Pharmaceutics 2026, 18(1), 130; https://doi.org/10.3390/pharmaceutics18010130 - 20 Jan 2026
Abstract
Modern medicine requires effective, continuous, and safe therapies, which largely depend on the targeted delivery and activity of the drug. This goal can be achieved by designing drug delivery systems with improved pharmacokinetic properties and enhanced drug transport to the affected tissue. Human [...] Read more.
Modern medicine requires effective, continuous, and safe therapies, which largely depend on the targeted delivery and activity of the drug. This goal can be achieved by designing drug delivery systems with improved pharmacokinetic properties and enhanced drug transport to the affected tissue. Human serum albumin (HSA) is an attractive carrier for the synthesis of therapeutic nanoparticles, several of which have already been approved by the United States Food and Drug Administration (FDA). The success of Abraxane as an effective treatment for metastatic breast cancer and non-small cell lung carcinoma, the application of Optison in ultrasound imaging, and the use of Nanocoll as an agent for SPECT diagnostics in sentinel node localisation confirm the strong potential of albumin-based systems. Further benefits are expected in patients with soft tissue cancers, as LadRx is seeking FDA marketing approval for Aldoxorubicin. The future of oncology lies in theranostics, which combines a tumour-localising factor on one platform with a drug targeting cancer cells and a factor that activates the cytotoxicity of the drug after it reaches the target tissue. This article presents recent advancements in albumin-based nanoparticles for drug delivery, targeting, and imaging. It also briefly discusses methods of synthesis and surface modification of albumin nanocarriers to enable targeted delivery to pathological sites. Finally, it outlines the latest approaches in multimodal theranostic platforms, highlighting albumin’s potential to improve cancer therapy. Full article
(This article belongs to the Special Issue Advanced Nanomaterials for Drug Delivery, 2nd Edition)
Show Figures

Figure 1

25 pages, 2813 KB  
Review
PSMA-Based Radiopharmaceuticals in Prostate Cancer Theranostics: Imaging, Clinical Advances, and Future Directions
by Ali Cahid Civelek
Cancers 2026, 18(2), 234; https://doi.org/10.3390/cancers18020234 - 12 Jan 2026
Viewed by 232
Abstract
Prostate cancer remains one of the most common malignancies in men worldwide, with incidence and mortality steadily increasing across diverse populations. While early detection and radical prostatectomy can achieve durable control in a subset of patients, approximately 40% of men will ultimately experience [...] Read more.
Prostate cancer remains one of the most common malignancies in men worldwide, with incidence and mortality steadily increasing across diverse populations. While early detection and radical prostatectomy can achieve durable control in a subset of patients, approximately 40% of men will ultimately experience biochemical recurrence often in the absence of clinically detectable disease. Conventional imaging approaches—CT, MRI, and bone scintigraphy—have limited sensitivity for early relapses, frequently leading to delayed diagnosis and suboptimal treatment planning. The discovery of prostate-specific membrane antigen (PSMA) in 1987 and its subsequent clinical translation into positron emission tomography (PET) imaging with [68Ga]Ga-PSMA-11 in 2012, followed by U.S. FDA approval in 2020, has transformed the landscape of prostate cancer imaging. PSMA PET has demonstrated superior accuracy over conventional imaging, as highlighted in the landmark proPSMA trial and now serves as the foundation for theranostic approaches that integrate diagnostic imaging with targeted radioligand therapy. The clinical approval of [177Lu]Lu-PSMA-617 (Pluvicto®: (lutetium Lu 177 vipivotide tetraxetan, Advanced Accelerator Applications USA, Inc., a Novartis company) has established targeted radioligand therapy as a viable option for men with metastatic castration-resistant prostate cancer, extending survival in patients with limited alternatives. Emerging strategies, including next-generation ligands with improved tumor uptake and altered clearance pathways, as well as the integration of artificial intelligence for imaging quantification, are poised to further refine patient selection, dosimetry, and treatment outcomes. This review highlights the evolution of PSMA-based imaging and therapy, discusses current clinical applications and limitations, and outlines future directions for optimizing theranostic strategies in prostate cancer care. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

27 pages, 1311 KB  
Review
Peptide-Functionalized Iron Oxide Nanoparticles for Cancer Therapy: Targeting Strategies, Mechanisms, and Translational Opportunities
by Andrey N. Kuskov, Lydia-Nefeli Thrapsanioti, Ekaterina Kukovyakina, Anne Yagolovich, Elizaveta Vlaskina, Petros Tzanakakis, Aikaterini Berdiaki and Dragana Nikitovic
Molecules 2026, 31(2), 236; https://doi.org/10.3390/molecules31020236 - 10 Jan 2026
Viewed by 359
Abstract
Therapeutic peptides have emerged as promising tools in oncology due to their high specificity, favorable safety profile, and capacity to target molecular hallmarks of cancer. Their clinical translation, however, remains limited by poor stability, rapid proteolytic degradation, and inefficient biodistribution. Iron oxide nanoparticles [...] Read more.
Therapeutic peptides have emerged as promising tools in oncology due to their high specificity, favorable safety profile, and capacity to target molecular hallmarks of cancer. Their clinical translation, however, remains limited by poor stability, rapid proteolytic degradation, and inefficient biodistribution. Iron oxide nanoparticles (IONPs) offer a compelling solution to these challenges. Owing to their biocompatibility, magnetic properties, and ability to serve as both drug carriers and imaging agents, IONPs have become a versatile platform for precision nanomedicine. The integration of peptides with IONPs has generated a new class of hybrid systems that combine the biological accuracy of peptide ligands with the multifunctionality of magnetic nanomaterials. Peptide functionalization enables selective tumor targeting and deeper tissue penetration, while the IONP core supports controlled delivery, MRI-based tracking, and activation of therapeutic mechanisms such as magnetic hyperthermia. These hybrids also influence the tumor microenvironment (TME), facilitating stromal remodeling and improved drug accessibility. Importantly, the iron-driven redox chemistry inherent to IONPs can trigger regulated cell death pathways, including ferroptosis and autophagy, inhibiting opportunities to overcome resistance in aggressive or refractory tumors. As advances in peptide engineering, nanotechnology, and artificial intelligence accelerate design and optimization, peptide–IONP conjugates are poised for translational progress. Their combined targeting precision, imaging capability, and therapeutic versatility position them as promising candidates for next-generation cancer theranostics. Full article
Show Figures

Graphical abstract

20 pages, 7392 KB  
Article
Anisotropic Gold Nanostars Functionalized with 2-Thiouracil: A Multifunctional Platform for Colorimetric Biosensing and Photothermal Cancer Therapy
by Tozivepi Aaron Munyayi, Anine Crous and Heidi Abrahamse
J. Nanotheranostics 2026, 7(1), 2; https://doi.org/10.3390/jnt7010002 - 8 Jan 2026
Viewed by 212
Abstract
This study presents a multifunctional theranostic platform based on anisotropic gold nanostars (AuNSs) functionalized with 2-thiouracil (2-TU) for cancer diagnostics and photothermal therapy (PTT). The unique plasmonic properties of AuNSs, combined with the anticancer and photothermal potential of 2-TU, were harnessed to create [...] Read more.
This study presents a multifunctional theranostic platform based on anisotropic gold nanostars (AuNSs) functionalized with 2-thiouracil (2-TU) for cancer diagnostics and photothermal therapy (PTT). The unique plasmonic properties of AuNSs, combined with the anticancer and photothermal potential of 2-TU, were harnessed to create a system capable of simultaneous colorimetric biosensing and therapeutic action. Under dual-wavelength irradiation (660 nm and 525 nm), the AuNSs–2-TU conjugate demonstrated enhanced photothermal conversion efficiency, selective cancer cell targeting, and signal amplification, resulting in a significant reduction in the IC50 for MCF-7 breast cancer cells. The system exhibited minimal cytotoxicity to normal fibroblasts (WS1), ensuring therapeutic precision. Compared to conventional spherical gold nanoparticles, this platform provides superior multifunctionality, including real-time biosensing with simple, naked-eye colorimetric readouts. These results highlight the potential of the AuNSs–2-TU conjugate as an innovative, minimally invasive nanotheranostic platform suitable for integrated cancer detection and treatment, particularly in resource-constrained settings. Full article
(This article belongs to the Special Issue Advances in Nanoscale Drug Delivery Technologies and Theranostics)
Show Figures

Graphical abstract

21 pages, 2602 KB  
Article
Identification of Key Sequence Motifs Essential for the Recognition of m6A Modification in RNA
by Aftab Mollah, Rushdhi Rauff, Sudeshi Abedeera, Chathurani Ekanayake, Chamali Thalagaha Mudiyanselage, Minhchau To, Helen Piontkivska and Sanjaya Abeysirigunawardena
Biomolecules 2026, 16(1), 97; https://doi.org/10.3390/biom16010097 - 7 Jan 2026
Viewed by 305
Abstract
N6-methyladenosine (m6A) constitutes the most prevalent nucleotide modification within eukaryotic messenger RNA (mRNA). Variations in m6A levels are associated with numerous human diseases and health conditions, including various forms of cancer, diabetes, neurological disorders, male infertility, and obesity. Nevertheless, [...] Read more.
N6-methyladenosine (m6A) constitutes the most prevalent nucleotide modification within eukaryotic messenger RNA (mRNA). Variations in m6A levels are associated with numerous human diseases and health conditions, including various forms of cancer, diabetes, neurological disorders, male infertility, and obesity. Nevertheless, the molecular mechanisms underpinning the recognition of m6A by different ‘reader’ proteins remain incompletely elucidated. In this study, we used phage display to identify key sequence features that methyl readers recognize in m6A. This study shows that m6A modifications affect the mRNA interactome. A peptide motif recognizing m6A in DRACH sequences suggests a common recognition mechanism, though proteins may use different methods to detect m6A in less accessible areas. The sequence of the hnRNP A1 RRM domain that aligns with the newly discovered m6A-binding peptide, m1p1, is crucial for the binding of m6A-modified RNAs, indicating a strong link between the m1p1 sequence and m6A recognition, which is key for recognizing m6A-modified, unstructured RNAs. Gaining a comprehensive understanding of the evolutionary influence of m6A on its reader proteins may facilitate the identification of additional m6A readers. These signature peptides could enhance theranostic approaches across cancers, enabling more targeted therapies. Full article
(This article belongs to the Section Molecular Biophysics: Structure, Dynamics, and Function)
Show Figures

Figure 1

21 pages, 687 KB  
Review
Radiolabelled FAPI Radiotracers in Oncology: A Comprehensive Review of Current Diagnostic and Emerging Therapeutic Applications
by Jolanta Czuczejko, Bogdan Małkowski, Jarosław Nuszkiewicz, Iga Hołyńska-Iwan, Paweł Waśniowski, Katarzyna Mądra-Gackowska, Wiktor Dróżdż and Karolina Szewczyk-Golec
Pharmaceuticals 2026, 19(1), 89; https://doi.org/10.3390/ph19010089 - 2 Jan 2026
Viewed by 430
Abstract
Background/Objectives: Fibroblast activation protein (FAP), which is abundantly expressed in cancer-associated fibroblasts (CAFs) across various epithelial malignancies, has emerged as a promising target for molecular imaging and radionuclide therapy. Although several reviews have addressed FAP-targeted diagnostics, a comprehensive synthesis integrating molecular biology, [...] Read more.
Background/Objectives: Fibroblast activation protein (FAP), which is abundantly expressed in cancer-associated fibroblasts (CAFs) across various epithelial malignancies, has emerged as a promising target for molecular imaging and radionuclide therapy. Although several reviews have addressed FAP-targeted diagnostics, a comprehensive synthesis integrating molecular biology, diagnostic performance, and early therapeutic development remains limited. This review summarises the current evidence on radionuclide-labelled FAP inhibitors (FAPIs), with particular emphasis on their diagnostic utility, emerging therapeutic applications, and the structural features that shape their biological behaviour. Methods: A structured literature search was conducted across PubMed, Scopus, and Web of Science, focusing on FAPI-based imaging and therapy. Results: Diagnostic studies consistently demonstrate high tumour-to-background contrast for [68Ga]Ga and [18F]-labelled FAPI radiotracers, particularly in tumours with prominent stromal components such as pancreatic, colorectal, breast, and head and neck cancers. FAPI PET/CT often surpasses [18F]FDG in lesion conspicuity in the brain, liver, and peritoneum. Therapeutic evidence shows encouraging tumour retention and safety profiles for agents such as [177Lu]Lu-FAP-2286 and [90Y]Y-FAPI-46, while α-emitting radiotracers (e.g., [225Ac]Ac-FAPI-04) demonstrate potent antitumor effects in preclinical models. Conclusions: Radiolabelled FAPI radiotracers hold significant potential as dual diagnostic and therapeutic agents, particularly for desmoplastic tumours with high CAF content. Nonetheless, clinical evidence remains in its early stages, and substantial questions persist regarding dosimetry, intertumoral variability in FAP expression, and optimal ligand selection for therapy. Continued development of next-generation FAPI constructs, along with well-designed prospective trials, will be crucial in defining the future role of FAPI-based theranostics in oncology. Full article
Show Figures

Graphical abstract

41 pages, 1227 KB  
Review
Lanthanide Nanotheranostics in Radiotherapy
by Shaofeng Han, Yurun Liu, Taoyang Cai, Yanru Liu and Shangjie Ge-Zhang
Int. J. Mol. Sci. 2026, 27(1), 426; https://doi.org/10.3390/ijms27010426 - 31 Dec 2025
Viewed by 489
Abstract
Radiotherapy, a cornerstone of cancer treatment, is critically limited by tumor radioresistance and off-target toxicity. Lanthanide-based nanomaterials (Ln-NPs) have recently emerged as a versatile and promising class of theranostic radiosensitizers to overcome these hurdles. This review comprehensively outlines the state-of-the-art in Ln-NP-enabled radiotherapy, [...] Read more.
Radiotherapy, a cornerstone of cancer treatment, is critically limited by tumor radioresistance and off-target toxicity. Lanthanide-based nanomaterials (Ln-NPs) have recently emerged as a versatile and promising class of theranostic radiosensitizers to overcome these hurdles. This review comprehensively outlines the state-of-the-art in Ln-NP-enabled radiotherapy, beginning with their fundamental physicochemical properties and synthesis and then delving into the multi-level mechanisms of radiosensitization, including high-Z element-mediated physical dose amplification, catalytic generation of reactive oxygen species (ROS), and disruption of DNA damage repair pathways. The unique capacity of certain Ln-NPs to serve as MRI contrast agents is highlighted as the foundation for image-guided, dose-painting radiotherapy. We critically summarize the preclinical and clinical progress of representative systems, benchmarking them against other high-Z nanomaterials. Finally, this work discusses the ongoing challenges, such as biocompatibility, targeted delivery, and regulatory hurdles, and envisages future directions, including combinatorial strategies with immunotherapy and the development of personalized nanotheranostic paradigms. Through this synthesis, this review aims to provide a clear roadmap for the continued development and clinical integration of lanthanide nanotheranostics in oncology. Full article
(This article belongs to the Special Issue New Advances in Radiopharmaceuticals and Radiotherapy)
Show Figures

Figure 1

47 pages, 10163 KB  
Review
Nanomedicine in Ovarian Cancer: Advances in Imaging, Targeted Delivery, and Theranostic Therapeutic Platforms
by Dorota Bartusik-Aebisher, Izabella Wilk and David Aebisher
Cancers 2026, 18(1), 86; https://doi.org/10.3390/cancers18010086 - 27 Dec 2025
Viewed by 610
Abstract
Ovarian cancer continues to be the most lethal gynaecological malignancy, principally due to its late-stage diagnosis, extensive peritoneal dissemination, chemoresistance, and limitations of current imaging and therapeutic strategies. By optimising pharmacokinetics, refining tumour-selective drug delivery, and supporting high-resolution, multimodal imaging, nanomedicine offers a [...] Read more.
Ovarian cancer continues to be the most lethal gynaecological malignancy, principally due to its late-stage diagnosis, extensive peritoneal dissemination, chemoresistance, and limitations of current imaging and therapeutic strategies. By optimising pharmacokinetics, refining tumour-selective drug delivery, and supporting high-resolution, multimodal imaging, nanomedicine offers a versatile platform to address these limitations. In this review, current progress across lipid-based, polymeric, inorganic, hybrid, and biomimetic nanocarriers is synthesised, emphasising how tailored physiochemical properties, surface functionalisation, and stimuli-responsive designs can improve tumour localisation, surmount stromal and ascetic barriers, and enable controlled drug release. Concurrently, significant advancement in imaging nanoprobes, including magnetic resonance imaging (MRI), positron emission tomography (PET)/single-photon emission computed tomography (SPECT), optical, near-infrared imaging (NIR), ultrasound, and photoacoustic systems, has evolved early lesion detection, intraoperative guidance, and quantitative monitoring of treatment. Diagnosis and therapy are further integrated within single platforms by emerging theranostic constructs, encouraging real-time visualisation of drug distribution and treatment response. Additionally, immune-nanomedicine, intraperitoneal depot systems, and nucleic acid-centred nanotherapies offer promising strategies to address immune suppression and molecular resistance in advanced ovarian cancer. In spite of noteworthy achievements, clinical translation is limited by complex manufacturing requirements, challenges with safety and stability, and restricted patient stratification. To unlock the full clinical potential of nanotechnology in ovarian cancer management, constant innovation in scalable design, regulatory standardisation, and integration of precision biomarkers will be necessary. Full article
(This article belongs to the Section Methods and Technologies Development)
Show Figures

Graphical abstract

39 pages, 3097 KB  
Review
Smart Cells Against Cancer: Advances in Cell-Based Drug Delivery and Diagnostics
by Lisa Gherardini, Giovanni Inzalaco, Sara Gargiulo, Lorenzo Franci and Monia Taranta
Pharmaceutics 2026, 18(1), 28; https://doi.org/10.3390/pharmaceutics18010028 - 25 Dec 2025
Viewed by 395
Abstract
Cell-based drug delivery has emerged as a powerful strategy to improve therapeutic targeting while reducing systemic toxicity. This approach is particularly valuable for anticancer agents, which are often limited by severe side effects arising from off-target activity and non-specific distribution. By using cells [...] Read more.
Cell-based drug delivery has emerged as a powerful strategy to improve therapeutic targeting while reducing systemic toxicity. This approach is particularly valuable for anticancer agents, which are often limited by severe side effects arising from off-target activity and non-specific distribution. By using cells as carriers, drugs can evade immune clearance, achieve prolonged circulation, and improve pharmacokinetic profiles, ultimately enhancing therapeutic efficacy. This review surveys the current landscape of cell-mediated drug delivery in oncology, emphasizing both fundamental principles and practical applications. We discuss the design and preparation of cellular carriers, examine the unique characteristics of commonly used cell types, and highlight recent technological innovations that are expanding their theranostic potential, focusing on strategies for delivery to challenging anatomical sites, with a dedicated focus on the brain. By consolidating recent advances and insights, this review aims to provide a comprehensive perspective on the promise and future directions of cell-based drug delivery for cancer therapy. Full article
(This article belongs to the Special Issue Cell-Mediated Delivery Systems)
Show Figures

Figure 1

17 pages, 2279 KB  
Article
L19-Conjugated Gold Nanoparticles for the Specific Targeting of EDB-Containing Fibronectin in Neuroblastoma
by Chiara Barisione, Silvia Ortona, Veronica Bensa, Caterina Ivaldo, Eleonora Ciampi, Simonetta Astigiano, Michele Cilli, Luciano Zardi, Mirco Ponzoni, Domenico Palombo, Giovanni Pratesi, Pier Francesco Ferrari and Fabio Pastorino
Pharmaceutics 2026, 18(1), 24; https://doi.org/10.3390/pharmaceutics18010024 - 24 Dec 2025
Viewed by 370
Abstract
Background/Objectives: Neuroblastoma (NB) is the most common extracranial solid tumor in children and accounts for 12–15% of pediatric cancer-related deaths. Current multimodal therapies lack specific cellular targets, causing systemic toxicity and drug resistance. The development of innovative tumor-targeted nanoformulations might represent a [...] Read more.
Background/Objectives: Neuroblastoma (NB) is the most common extracranial solid tumor in children and accounts for 12–15% of pediatric cancer-related deaths. Current multimodal therapies lack specific cellular targets, causing systemic toxicity and drug resistance. The development of innovative tumor-targeted nanoformulations might represent a promising approach to enhance NB diagnosis and antitumor efficacy, while decreasing off targets side effects. Fibronectin extra-domain B (FN-EDB) is upregulated in the tumor microenvironment. Methods: FN-EDB expression was evaluated by immunohistochemical staining in cell line-derived and tumor patient-derived animal models of NB. A gold nanoparticle, decorated with an antibody (Ab) recognizing FN-EDB (L19-AuNP) was developed by the company Nano Flow and its tumor binding was tested by ELISA in vitro and in patient-derived xenograft (PDX) models of NB by photoacoustic imaging in vivo. Results: All animal models of NB used have been shown to express FN-EDB. L19 Ab demonstrated excellent binding specificity to FN-EDB both when used in free form and after conjugation to AuNP. Compared to the non-functionalized (no Ab L19-coupled) AuNP, which showed an increase in PDI and zeta potential over time, making them unsuitable for use in in vivo studies, L19-AuNP demonstrated good stability. In vivo, L19-AuNP specifically homed into PDX models of NB, accumulating better in tumors expressing higher levels of FN-EDB. Negligible distribution to healthy organs occurred. Conclusions: In this preliminary study, L19-AuNP was shown to be a novel diagnostic tool specifically for binding NB expressing FN-EDB, paving the way for the development of theranostic nanoformulations co-encapsulating gold moiety and standard-of-care therapy for NB. Full article
(This article belongs to the Special Issue Nanomedicine and Nanotechnology: Recent Advances and Applications)
Show Figures

Graphical abstract

19 pages, 2482 KB  
Review
Application of Metal-Doped Nanomaterials in Cancer Diagnosis and Treatment
by Xinhao Jin and Qi Sun
J. Nanotheranostics 2025, 6(4), 35; https://doi.org/10.3390/jnt6040035 - 17 Dec 2025
Viewed by 454
Abstract
Cancer remains a severe global health threat, with traditional therapies often plagued by limited efficacy and significant side effects. The emergence of nanotechnology, particularly metal-doped nanomaterials, offers a promising avenue for integrating diagnostic and therapeutic functions into a single platform, enabling a theranostic [...] Read more.
Cancer remains a severe global health threat, with traditional therapies often plagued by limited efficacy and significant side effects. The emergence of nanotechnology, particularly metal-doped nanomaterials, offers a promising avenue for integrating diagnostic and therapeutic functions into a single platform, enabling a theranostic approach to oncology. This article explores the design and application of various metal-doped nanosystems, including gadolinium-doped selenium molybdenum nanosheets for magnetic resonance/photoacoustic dual-mode imaging and photothermal therapy, and metal-doped hollow mesoporous silica nanoparticles that leverage the tumor’s acidic microenvironment to release ions for catalytic generation of reactive oxygen species. Despite their promise, the limited enzyme-like activity of some nanozymes, insufficient endogenous hydrogen peroxide in tumors, and the tumor microenvironment’s defensive mechanisms, such as high glutathione levels, can restrict therapeutic efficacy. Looking forward, the outlook for the field is contingent upon advancing material engineering strategies. Future research should prioritize the development of intelligent, multifunctional nanoplatforms that can dynamically respond to and remodel the tumor microenvironment. Innovations in surface modification for enhanced targeting, alongside rigorous preclinical studies focused on safety and standardized manufacturing, are crucial for bridging the gap between laboratory research and clinical application, ultimately paving the way for personalized cancer medicine. Full article
(This article belongs to the Special Issue Feature Review Papers in Nanotheranostics)
Show Figures

Figure 1

38 pages, 2034 KB  
Review
The Application of Nanomaterials in Breast Cancer
by Kexin Guo, Yue Sun and Huihua Xiong
Pharmaceutics 2025, 17(12), 1608; https://doi.org/10.3390/pharmaceutics17121608 - 14 Dec 2025
Viewed by 553
Abstract
Breast cancer is one of the most prevalent malignant tumors worldwide, with the highest incidence and mortality among women. Early precise diagnosis and the development of efficient treatment regimens remain major clinical challenges. Harnessing the programmable size, surface chemistry, and tumor microenvironment (TME) [...] Read more.
Breast cancer is one of the most prevalent malignant tumors worldwide, with the highest incidence and mortality among women. Early precise diagnosis and the development of efficient treatment regimens remain major clinical challenges. Harnessing the programmable size, surface chemistry, and tumor microenvironment (TME) responsiveness of nanomaterials, there is tremendous potential for their applications in breast cancer diagnosis and therapy. In the diagnostic arena, nanomaterials serve as core components of novel contrast agents (e.g., gold nanorods, quantum dots, superparamagnetic iron oxide nanoparticles) and biosensing platforms, substantially enhancing the sensitivity and specificity of molecular imaging modalities—such as magnetic resonance imaging (MRI), computed tomography (CT), and fluorescence imaging (FLI)—and enabling high-sensitivity detection of circulating tumor cells and tumor-derived exosomes, among various liquid biopsy biomarkers. In therapy, nanoscale carriers (e.g., liposomes, polymeric micelles) improve tumor targeting and accumulation efficiency through passive and active targeting strategies, thereby augmenting anticancer efficacy while effectively reducing systemic toxicity. Furthermore, nanotechnology has spurred the rapid advancement of emerging modalities, including photothermal therapy (PTT), photodynamic therapy (PDT), and immunotherapy. Notably, the construction of theranostic platforms that integrate diagnostic and therapeutic units within a single nanosystem enables in vivo, real-time visualization of drug delivery, treatment monitoring, and therapeutic response feedback, providing a powerful toolkit for advancing breast cancer toward personalized, precision medicine. Despite challenges that remain before clinical translation—such as biocompatibility, scalable manufacturing, and standardized evaluation—nanomaterials are undoubtedly reshaping the paradigm of breast cancer diagnosis and treatment. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Graphical abstract

25 pages, 359 KB  
Review
The Gastrointestinal Tract: A Unique Battlefield for Bioengineering Delivery Platforms
by Teng Ma and Siyu Sun
Bioengineering 2025, 12(12), 1347; https://doi.org/10.3390/bioengineering12121347 - 10 Dec 2025
Viewed by 612
Abstract
Traditional drug delivery methods for gastrointestinal diseases, including oral and systemic administration, often suffer from degradation, inadequate mucosal absorption, and off-target toxicity. Consequently, these methods result in low bioavailability and suboptimal therapeutic outcomes for localized conditions such as inflammation and early-stage cancer. This [...] Read more.
Traditional drug delivery methods for gastrointestinal diseases, including oral and systemic administration, often suffer from degradation, inadequate mucosal absorption, and off-target toxicity. Consequently, these methods result in low bioavailability and suboptimal therapeutic outcomes for localized conditions such as inflammation and early-stage cancer. This review examines the innovative integration of advanced bioengineering platforms with therapeutic gastrointestinal endoscopy to address these delivery challenges. We concentrate on three principal bioengineered platforms: (1) nanoparticle systems (e.g., lipid, polymeric, and inorganic nanoparticles) designed for localized chemotherapy and theranostics; (2) in situ-forming hydrogels that serve as intelligent wound management materials and sustained drug depots; and (3) drug-eluting and biodegradable stents that convert passive luminal scaffolds into active, long-term drug-releasing devices. An analysis of these platforms demonstrates that their synergy with endoscopy facilitates precise, minimally invasive, and sustained local therapy, potentially transforming the treatment landscape for gastrointestinal diseases such as cancer and inflammatory bowel disease. Additionally, we investigate advanced strategies, including active targeting and stimulus-responsive release mechanisms, to enhance spatial precision. Despite promising preclinical advancements, clinical translation encounters challenges related to long-term biocompatibility, scalable manufacturing, regulatory pathways for drug-device combinations, and cost-effectiveness. Ultimately, the convergence of bioengineering and endoscopy presents significant potential to usher in a new era of precise, localized, and sustained micro-invasive treatments in gastroenterology. Full article
(This article belongs to the Special Issue Bioengineering Platforms for Drug Delivery)
Show Figures

Graphical abstract

42 pages, 2995 KB  
Review
Plasma Cell Myeloma: Biochemical Insights into Diagnosis, Treatment, and Smart Nanocarrier-Based Therapeutic Development
by Lizeth Geraldine Muñoz, Sixta Palencia Luna and Andrés Felipe Chamorro
Pharmaceutics 2025, 17(12), 1570; https://doi.org/10.3390/pharmaceutics17121570 - 5 Dec 2025
Viewed by 656
Abstract
Plasma cell myeloma (PCM) is classified as a blood cancer and is characterized by the abnormal proliferation of plasma cells in the bone marrow and the excessive production of monoclonal immunoglobulins, which lead to permanent damage to vital organs. Although treatment strategies have [...] Read more.
Plasma cell myeloma (PCM) is classified as a blood cancer and is characterized by the abnormal proliferation of plasma cells in the bone marrow and the excessive production of monoclonal immunoglobulins, which lead to permanent damage to vital organs. Although treatment strategies have improved with the development of proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and monoclonal antibodies (mAbs), PCM remains an incurable disease due to its molecular heterogeneity and the development of drug resistance. In this review, we discuss the biochemical and molecular foundations underlying the diagnosis and treatment of PCM, emphasizing both traditional and advanced approaches. Classical methods such as serum protein electrophoresis (SPEP), immunofixation electrophoresis (IFE), and serum free light chain (sFLC) determination are highlighted alongside their integration with highly sensitive techniques like mass spectrometry (MS) and next-generation sequencing (NGS). Special attention is given to nanotechnology-based systems, including liposomes, polymeric nanoparticles (NPs), dendrimers, and hybrid nanocapsules, which enable controlled drug release, targeted delivery, and the minimization of systemic toxicity. Increasingly, nanomaterials are being shown to greatly enhance the biodistribution and pharmacokinetics of anticancer drugs, leading to improved therapeutic effects and escaping resistance mechanisms by employing multifunctional strategies that include dual drug co-encapsulation, pH-sensitive release and theranostic applications. Furthermore, the integration of nanotechnology with immunotherapy platforms represents a paradigm shift toward precision and personalized medicine for the treatment of PCM. Overall, this review views nanotechnology as an enabling technology to improve therapeutic effectiveness, minimize toxicity and open new avenues toward next-generation smart and personalized therapeutics for the treatment of PCM. Full article
(This article belongs to the Special Issue Nanomedicine and Nanotechnology: Recent Advances and Applications)
Show Figures

Figure 1

38 pages, 1179 KB  
Review
Therapeutic Advances in Metastatic Prostate Cancer: A Journey from Standard of Care to New Emerging Treatment
by Rossella Cicchetti, Martina Basconi, Giulio Litterio, Angelo Orsini, Marco Mascitti, Alessio Digiacomo, Gaetano Salzano, Octavian Sabin Tătaru, Matteo Ferro, Carlo Giulioni, Angelo Cafarelli, Luigi Schips and Michele Marchioni
Int. J. Mol. Sci. 2025, 26(23), 11665; https://doi.org/10.3390/ijms262311665 - 2 Dec 2025
Viewed by 2906
Abstract
Prostate cancer (PCa) remains one of the most prevalent malignancies among men worldwide and continues to pose significant therapeutic challenges, especially in its metastatic and castration-resistant forms. Over the past two decades, the treatment paradigm has evolved from monotherapy with androgen deprivation therapy [...] Read more.
Prostate cancer (PCa) remains one of the most prevalent malignancies among men worldwide and continues to pose significant therapeutic challenges, especially in its metastatic and castration-resistant forms. Over the past two decades, the treatment paradigm has evolved from monotherapy with androgen deprivation therapy (ADT) to a multifaceted approach integrating chemotherapy, androgen receptor axis-targeted therapies (ARATs), radiopharmaceuticals, and precision medicine. This review explores the molecular underpinnings of PCa, including genetic and epigenetic alterations such as BRCA1/2, TP53, and PTEN mutations, and their role in disease progression and treatment resistance. We detail the evidence supporting the integration of systemic agents like abiraterone, enzalutamide, and darolutamide into both hormone-sensitive and castration-resistant settings. Furthermore, we highlight the expanding role of radioligand therapies, including radium-223 and Lutetium-177-labeled PSMA-617 (Lu-PSMA-617), as well as the growing impact of PARP inhibitors in genomically selected patients. The emergence of theranostic strategies and next-generation sequencing has paved the way for personalized treatment algorithms, moving toward a truly precision oncology model in PCa. This comprehensive review synthesizes current therapeutic strategies, clinical trial evidence, and future directions aimed at optimizing outcomes and quality of life for patients with advanced prostate cancer. Full article
Show Figures

Figure 1

Back to TopTop