1
|
Lan X, Johnston E, Ning T, Chen G, Haglund L, Li J. Immunomodulatory bioadhesive technologies. Biomaterials 2025; 321:123274. [PMID: 40156979 DOI: 10.1016/j.biomaterials.2025.123274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Bioadhesives have found significant use in medicine and engineering, particularly for wound care, tissue engineering, and surgical applications. Compared to traditional wound closure methods such as sutures and staples, bioadhesives offer advantages, including reduced tissue damage, enhanced healing, and ease of implementation. Recent progress highlights the synergy of bioadhesives and immunoengineering strategies, leading to immunomodulatory bioadhesives capable of modulating immune responses at local sites where bioadhesives are applied. They foster favorable therapeutic outcomes such as reduced inflammation in wounds and implants or enhanced local immune responses to improve cancer therapy efficacy. The dual functionalities of bioadhesion and immunomodulation benefit wound management, tissue regeneration, implantable medical devices, and post-surgical cancer management. This review delves into the interplay between bioadhesion and immunomodulation, highlighting the mechanobiological coupling involved. Key areas of focus include the modulation of immune responses through chemical and physical strategies, as well as the application of these bioadhesives in wound healing and cancer treatment. Discussed are remaining challenges such as achieving long-term stability and effectiveness, necessitating further research to fully harness the clinical potential of immunomodulatory bioadhesives.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Evan Johnston
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Tianqin Ning
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Pine Ave W, Montreal, Quebec, H3A 1A3, Canada
| | - Lisbet Haglund
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Shriners Hospital for Children, 1003 Decarie Blvd, Montreal, Quebec, H4A 0A9, Canada.
| | - Jianyu Li
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada.
| |
Collapse
|
2
|
Gong Z, Zhou D, Wu D, Han Y, Yu H, Shen H, Feng W, Hou L, Chen Y, Xu T. Challenges and material innovations in drug delivery to central nervous system tumors. Biomaterials 2025; 319:123180. [PMID: 39985979 DOI: 10.1016/j.biomaterials.2025.123180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Central nervous system (CNS) tumors, encompassing a diverse array of neoplasms in the brain and spinal cord, pose significant therapeutic challenges due to their intricate anatomy and the protective presence of the blood-brain barrier (BBB). The primary treatment obstacle is the effective delivery of therapeutics to the tumor site, which is hindered by multiple physiological, biological, and technical barriers, including the BBB. This comprehensive review highlights recent advancements in material science and nanotechnology aimed at surmounting these delivery challenges, with a focus on the development and application of nanomaterials. Nanomaterials emerge as potent tools in designing innovative drug delivery systems that demonstrate the potential to overcome the limitations posed by CNS tumors. The review delves into various strategies, including the use of lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, all of which are engineered to enhance drug stability, BBB penetration, and targeted tumor delivery. Additionally, this review highlights the burgeoning role of theranostic nanoparticles, integrating therapeutic and diagnostic functionalities to optimize treatment efficacy. The exploration extends to biocompatible materials like biodegradable polymers, liposomes, and advanced material-integrated delivery systems such as implantable drug-eluting devices and microfabricated devices. Despite promising preclinical results, the translation of these material-based strategies into clinical practice necessitates further research and optimization.
Collapse
Affiliation(s)
- Zhenyu Gong
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China; Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Dairan Zhou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, 230601, PR China
| | - Yaguang Han
- Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Hao Yu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, PR China
| | - Haotian Shen
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Lijun Hou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| | - Tao Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
3
|
Martin JD, Toh K, Martin MR, Chen P, Wang C, Igarashi K, Mpekris F, Stylianopoulos T, Stuber MD, Kataoka K, Cabral H. Bone marrow vessels are hyperpermeable to macromolecules and nanoscale medicine in a size-dependent manner. J Control Release 2025; 382:113669. [PMID: 40158811 DOI: 10.1016/j.jconrel.2025.113669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/22/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Bone marrow (BM) has roles in health and disease, so systemically administered nanocarriers (NCs) targeting or avoiding BM are desirable. While the hydrodynamic diameter of NCs can be tuned to target or avoid various organs, the size dependence of extravasation from BM vessels is unknown. To clarify the size dependence of passive transvascular transport in the BM, we performed vessel permeability measurements in murine calvaria using confocal fluorescent microscopy with fluorescently labeled dextrans, albumin, and polymeric micelles as model probes. Unexpectedly, we found the permeability of BM vessels to macromolecules decreases with increasing hydrodynamic diameter between 4 nm and 32 nm. We modeled this permeability data with mathematical models to predict an effective pore size for sinusoids of 47 nm and non-sinusoids of 37 nm, with estimated maximum pore sizes of 61 nm and 53 nm, respectively. Finally, we tested these model predictions by demonstrating that the extravasation of 70 nm polymeric micelles, which are larger than the estimated maximum pore size, is hindered relative to 30 nm polymeric micelles. These results establish design criteria for controlling NC hydrodynamic diameter towards modulating delivery to BM.
Collapse
Affiliation(s)
- John D Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, Japan
| | - Margaret R Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Chenyu Wang
- Process Systems and Operations Research Laboratory, Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA
| | - Kazunori Igarashi
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Matthew D Stuber
- Process Systems and Operations Research Laboratory, Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA.
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan.
| |
Collapse
|
4
|
Gabizon AA. Cancer nanomedicine from a clinician-scientist perspective: Lessons and prospects. J Control Release 2025; 382:113731. [PMID: 40228664 DOI: 10.1016/j.jconrel.2025.113731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
The nanomedicine field has progressed enormously in the last couple of decades. From a loose group of liposomologists, polymer scientists, chemical engineers, and experts in metal nanoparticles, mesoporous silica, and other nanomaterials, the field has gradually consolidated and has generated vast amounts of research and clinical data, but, until the development of lipid nanoparticle (LNP)-based vaccinations for Covid-19, has remained with low visibility in the clinic. Applications in the cancer field are the most frequently sought projects in nanomedicine. For the last 45 years, my clinical career has mingled with my research career focusing on ways to formulate drugs in liposomes to improve their safety and efficacy in cancer therapy. In this review, I will discuss my contribution to the development of pegylated liposomal doxorubicin and other cancer nanomedicines from my privileged position as a clinician and scientist.
Collapse
Affiliation(s)
- Alberto A Gabizon
- The Leah and Jakub Susskind Nano-Oncology Research Laboratory at the Helmsley Cancer Center, Shaare Zedek Medical Center and the Hebrew University-Faculty of Medicine, Jerusalem, Israel.
| |
Collapse
|
5
|
Neophytou C, Charalambous A, Voutouri C, Angeli S, Panagi M, Stylianopoulos T, Mpekris F. Sonopermeation combined with stroma normalization enables complete cure using nano-immunotherapy in murine breast tumors. J Control Release 2025; 382:113722. [PMID: 40233830 PMCID: PMC12076078 DOI: 10.1016/j.jconrel.2025.113722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
Nano-immunotherapy shows great promise in improving patient outcomes, as seen in advanced triple-negative breast cancer, but it does not cure the disease, with median survival under two years. Therefore, understanding resistance mechanisms and developing strategies to enhance its effectiveness in breast cancer is crucial. A key resistance mechanism is the pronounced desmoplasia in the tumor microenvironment, which leads to dysfunction of tumor blood vessels and thus, to hypoperfusion, limited drug delivery and hypoxia. Ultrasound sonopermeation and agents that normalize the tumor stroma have been employed separately to restore vascular abnormalities in tumors with some success. Here, we performed in vivo studies in two murine, orthotopic breast tumor models to explore if combination of ultrasound sonopermeation with a stroma normalization drug can synergistically improve tumor perfusion and enhance the efficacy of nano-immunotherapy. We found that the proposed combinatorial treatment can drastically reduce primary tumor growth and in many cases tumors were no longer measurable. Overall survival studies showed that all mice that received the combination treatment survived and rechallenge experiments revealed that the survivors obtained immunological memory. Employing ultrasound elastography and contrast enhanced ultrasound along with proteomics analysis, flow cytometry and immunofluorescene staining, we found the combinatorial treatment reduced tumor stiffness to normal levels, restoring tumor perfusion and oxygenation. Furthermore, it increased infiltration and activity of immune cells and altered the levels of immunosupportive chemokines. Finally, using machine learning analysis, we identified that tumor stiffness, CD8+ T cells and M2-type macrophages were strong predictors of treatment response.
Collapse
Affiliation(s)
- Constantina Neophytou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Stella Angeli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.
| |
Collapse
|
6
|
Wang Y, Yan X, Lu D, Wang H, Li K, Chen L, Wang L, Cheng S, Wang G, Wang R, Xue Y, Li Y, Xie R, Ding L. Glycan Covalent Targeting Chimera-Based T-Cell Glycoengineering for Boosting Immunotherapy. Chemistry 2025; 31:e202500364. [PMID: 40207574 DOI: 10.1002/chem.202500364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/21/2025] [Accepted: 04/10/2025] [Indexed: 04/11/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is one of the most effective approaches in cancer immunotherapy. However, shortcomings such as loss of target antigens and poor infiltration remain in the treatment of solid tumors. Herein, we propose an approach to glycoengineer T cells based on glycan covalent targeting chimera (gcTAC), to enhance immunotherapy by modulating the glycan recognition behavior of T cells. We select hydrazide-modified phenylboronic acid (PBA) as gcTAC. The hydrazide group can covalently couple with the aldehyde group generated by the oxidation of galactose/N-acetylgalactosamine on the surface of T cells, while the PBA at the other end can specifically bind to sialic acids (Sia) of tumor cells, thereby enhancing the killing effect of T cells. At the same time, T cells covalently bound to tumor cells surfaces act as a blockade of Sia sites, which can disrupt the recognition between Siglec on natural killer (NK) cells and Sia on tumor cells, further enhancing the immune-killing effect of combined T-NK cell therapy. Our approach represents a novel concept to promote immune killing through cascading interventions in T-tumor and tumor-NK intercellular glycan recognition, thus providing a solution to the problem of immune escape in cancer therapy.
Collapse
Affiliation(s)
- Yichun Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xiaomin Yan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Di Lu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Haiqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ke Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Lan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Songtao Cheng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ruiyuan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yadong Xue
- Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365 East Renmin Road, Jinhua, 321000, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ran Xie
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
7
|
Wang S, Cao R, Lin Z, He W, Liao H, Xu J, Guo Y. Biocompatible Astragalus polysaccharide-based nanogels for oncology: synthesis, characterization, and therapeutic potential. Int J Biol Macromol 2025:144544. [PMID: 40409659 DOI: 10.1016/j.ijbiomac.2025.144544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/12/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
In recent years, drug delivery systems have become an important platform for cancer diagnosis and treatment. However, developing a novel nanoplatform that synergistically integrates multimodal therapies for precision tumor treatment remains a significant challenge. To address this, we employed Astragalus polysaccharide (ASP) with immunomodulatory function as the framework to load podophyllotoxin (PPT) and IR780, and constructed a novel ROS/GSH/NIR triple-response nanoplatform (IR780@PPTASP). Under simulated tumor microenvironment conditions, IR780@PPTASP underwent controlled collapse and drug release. The loaded IR780 photosensitizer endowed IR780@PPTASP with exceptional photothermal and photodynamic properties. To further investigate the molecular mechanism of IR780@PPTASP on A549 cells, we conducted a series of experiments. The results demonstrated that IR780@PPTASP could significantly inhibit A549 cell proliferation and migration, induce the production of reactive oxygen species (ROS), and promote A549 cell apoptosis. Furthermore, in vivo experiments revealed that IR780@PPTASP could effectively inhibit the proliferation and metastasis of zebrafish transplanted tumors. Notably, IR780@PPTASP promoted the release of nitric oxide (NO), increased the expression of CD40, CD80, CD86, and MHC-II, thereby promoting macrophage maturation and activating T cell function. The developed ROS/GSH/NIR triple-response nanoplatform enables a synergistic approach to chemo-phototherapy-immunotherapy, offering significant promise for the precision treatment of tumors, effectively achieving multiple therapeutic effects simultaneously.
Collapse
Affiliation(s)
- Sibei Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China
| | - Ruyu Cao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China
| | - Zhen Lin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China
| | - Wenrui He
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China
| | - Haibing Liao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jing Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China.
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China.
| |
Collapse
|
8
|
Gutierrez-Sandoval R, Gutiérrez-Castro F, Muñoz-Godoy N, Rivadeneira I, Sobarzo A, Iturra J, Krakowiak F, Alarcón L, Dorado W, Lagos A, Montenegro D, Muñoz I, Aguilera R, Toledo A. Beyond Exosomes: An Ultrapurified Phospholipoproteic Complex (PLPC) as a Scalable Immunomodulatory Platform for Reprogramming Immune Suppression in Metastatic Cancer. Cancers (Basel) 2025; 17:1658. [PMID: 40427155 DOI: 10.3390/cancers17101658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Dendritic-cell-derived exosomes (DEXs) have demonstrated immunostimulatory potential in cancer immunotherapy, yet their clinical application remains constrained by their cryodependence, compositional heterogeneity, and limited scalability. To address these limitations, we developed an ultrapurified phospholipoproteic complex (PLPC), a dendritic-secretome-derived formulation stabilized through ultracentrifugation and lyophilization that has been engineered to preserve its immunological function and structural integrity. Methods: Secretomes were processed under four conditions (fresh, concentrated, cryopreserved, and lyophilized PLPC) and compared through proteomic and functional profiling. Mass spectrometry (LC-MS/MS) analysis revealed that the PLPC retained a significantly enriched set of immunoregulatory proteins-including QSOX1, CCL22, and SDCBP-and exhibited superior preservation of post-translational modifications. Results: Ex vivo co-culture assays with human peripheral blood mononuclear cells (PBMCs) demonstrated that the PLPC induced robust secretion of IFN-γ, TNF-α, and IL-6 while concurrently suppressing IL-10, achieving an IFN-γ/IL-10 ratio exceeding 3.5. Flow cytometry confirmed the substantial activation of both CD4⁺ and CD8⁺ T cells, while apoptosis assays showed selective tumor cytotoxicity (>55% tumor apoptosis) with minimal impact on non-malignant cells (>92% viability). Conclusions: These findings establish the PLPC as a reproducible, Th1-polarizing immunomodulator with selective antitumor activity, ambient-temperature stability, and compatibility with non-invasive administration. Overall, the PLPC emerges as a scalable, cell-free immunotherapeutic platform with translational potential to reprogram immune suppression in metastatic therapy-resistant cancer settings.
Collapse
Affiliation(s)
| | | | - Natalia Muñoz-Godoy
- Department of Cancer Research, Flowinmunocell-Bioexocell Group, 08028 Barcelona, Spain
| | - Ider Rivadeneira
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Adolay Sobarzo
- Department of Biological and Chemistry Sciences, Faculty of Medicine and Science, San Sebastian University, Concepción 4080871, Chile
| | - Jordan Iturra
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Francisco Krakowiak
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
- Department of Molecular Oncopathology, Bioclas, Concepción 4030000, Chile
| | - Luis Alarcón
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Wilson Dorado
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Andy Lagos
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Diego Montenegro
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Ignacio Muñoz
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Rodrigo Aguilera
- Department of Outreach and Engagement Programs, OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Andres Toledo
- Department of Oncopathology, OGRD Alliance, Lewes, DE 19958, USA
| |
Collapse
|
9
|
Yang H, Li R, Jin S, Tian Y, Wang C, Sun Y, Dai Z, Cheng W. Targeted nanosensitizer-augmented sono-immunotherapy with STING agonist to remodel the immune microenvironment in hepatocellular carcinoma. Acta Biomater 2025:S1742-7061(25)00354-X. [PMID: 40349900 DOI: 10.1016/j.actbio.2025.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/22/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignant disease of the liver. Although immunotherapy offers new opportunities for treating advanced HCC, its therapeutic effect is still limited by the immunosuppressive tumor microenvironment (TME). Herein, a nanosensitizer RGD@Ce6@MSA-2@Liposome (RCM-Lip) is synthesized to specifically initiate the HCC tumor immune microenvironment through sonodynamic therapy (SDT)-triggered immunogenic cell death (ICD) and MSA-2-activated cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. RCM-Lip consists of a sonosensitizer (Chlorin e6, Ce6) with a STING agonist (MSA-2) and a tumor targeting peptide RGD inserted on the outer liposome surface. Under ultrasound irradiation, RCM-Lip generates reactive oxygen species that induce cytotoxicity and apoptosis of tumor cells. Meanwhile, tumor antigens released by apoptosis are taken up by dendritic cells (DCs), while STING is activated in the DCs by MSA-2. Moreover, DC maturation is stimulated, further enhancing the systematic anti-tumor immune responses. Sono-immunotherapy mediated by RCM-Lip promotes DCs maturation and tumor infiltration of CD8+T cells, increasing inflammatory cytokine secretion. Consequently, the immunologically "cold" TME of HCC is successfully turned into a "hot" one, leading to a significant tumor suppression effect with good bio-safety. These results suggest a promising method for precise tumor targeting and synergistic cancer sono-immunotherapy. STATEMENT OF SIGNIFICANCE: Our study addressed the therapeutic dilemma of hepatocellular carcinoma (HCC) as an immunological "cold" tumor by the synergistic application of sonodynamic therapy (SDT) and STING agonist. The cGAS-STING signaling pathway plays a pivotal role in innate immunity against cancer, but the clinical application of STING agonists were hampered by inflammatory responses due to off-target activation. Our innovative solution introduces RGD-targeted peptide to encapsulate sonosensitizer and STING agonist, strengthening therapeutic effects and reducing systemic toxicity. The targeted sono-immunotherapy promoted DCs maturation and tumor infiltration of CD8+T cells, producing intense tumor-killing effect on mice model with good bio-safety. As a result, the immunological "cold" tumor microenvironment of HCC is successfully turned into a "hot" one.
Collapse
Affiliation(s)
- Huajing Yang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Rui Li
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Shiyang Jin
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Yucao Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China.
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China.
| |
Collapse
|
10
|
Minopoli A, Perini G, Cui L, Palmieri V, De Spirito M, Papi M. Biomaterial-driven 3D scaffolds for immune cell expansion toward personalized immunotherapy. Acta Biomater 2025:S1742-7061(25)00351-4. [PMID: 40348072 DOI: 10.1016/j.actbio.2025.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/12/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Immunotherapy has emerged as a transformative medical approach in recent years, providing novel treatments for cancer eradication, autoimmune disorders, and infectious diseases. Fundamental to the success of therapy is the enrichment of the immune cell population, particularly T cells, natural killer cells, and dendritic cells. However, achieving a robust and long-term proliferation of immune cells is still challenging both in vivo and ex vivo. In vivo expansion leverages the patient's natural microenvironment and regulatory mechanisms through therapeutic interventions like immune checkpoint inhibitors, cytokine therapy, and targeted antibodies. This approach fosters long-term immune memory and sustained protection. In contrast, ex vivo expansion involves isolation, manipulation, and expansion of the immune cells under controlled conditions before reinfusion, allowing for precise control over the process and generating potent immune cell populations. Hydrogels, due to their tunable biomechanical properties, high biocompatibility, and ability to mimic the extracellular matrix, provide an ideal platform for both in vivo and ex vivo immune cell expansion. For instance, hydrogel-based scaffolds or beads can facilitate a controlled and efficient expansion of immune cells ex vivo, whereas injectable and implantable hydrogels can provide innovative solutions for enhancing immune cell activity within the patient supporting prolonged immune cell activity. This review aims to elucidate the importance of hydrogel-based strategies in immune cell expansion, advancing the development of effective, personalized immunotherapies to improve patient outcomes. STATEMENT OF SIGNIFICANCE: This review highlights the transformative potential of hydrogel-based 3D scaffolds in advancing personalized immunotherapy. By integrating in vivo and ex vivo strategies, hydrogels provide an innovative platform to enhance immune cell expansion, addressing critical challenges in immunotherapy. The discussion emphasizes the unique biomechanical and biochemical tunability of hydrogels, enabling precise mimicry of the extracellular matrix to support T cell proliferation, activation, and memory formation. These advances offer scalable, cost-effective solutions for producing high-quality immune cells, contributing to more effective cancer treatments, autoimmune disease management, and infectious disease control. By bridging materials science and immunology, this work underscores the pivotal role of hydrogels in shaping the future of immune-based therapies.
Collapse
Affiliation(s)
- Antonio Minopoli
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Lishan Cui
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Valentina Palmieri
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy; Istituto dei Sistemi Complessi, Consiglio Nazionale delle Ricerche, CNR, via dei Taurini 19, 00185 Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy.
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy.
| |
Collapse
|
11
|
Chu Y, Wang L, Huang X, Lao C, Luo L, Chen J, Jin F, Zhao L, Hu K, Lv M, Li X, Yang Y, Zhao W, Wang W. Virus-mimicking nanoparticle potentiates in-situ cancer vaccines by reversing intratumoral DCs via stimulating cytosolic nucleic acid sensors. J Control Release 2025; 383:113801. [PMID: 40339656 DOI: 10.1016/j.jconrel.2025.113801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/17/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
The major challenge for in-situ cancer vaccines is to activate the dendritic cells (DCs) within the immunosuppressive tumor to initiate tumor-specific immune responses. By contrast, oncolytic viruses can always activate intratumoral DCs to initiate virus-specific immunity to eliminate viruses which will in turn reduce the anti-tumor effect of these viruses. To bridge the two approaches, we developed virus-mimicking nanoparticle (V-mimic) through self-assembly of artificial cyclic gadolinium-based GAMP (c·Gd·GAMP, a STING agonist), double-stranded RNA analog (Poly I:C), and indocyanine green (ICG). Within the V-mimic, c·Gd·GAMP and Poly I:C can simulate the DNA and RNA produced by virus replication to reverse the inhibition of the immunosuppressive tumor microenvironment on DCs by promoting the IFN-β secreting via activating nucleic acid sensors, thereby eliciting anti-tumor immune response in a manner akin to the immune response triggered by viruses. Notably, the V-mimic significantly promoted IFN-β secretion, enhanced DCs maturation, and elicited robust tumor-specific immune memory when combined with photothermal therapy. In multiple tumor models, significant tumor inhibition was observed, including a metastasis and recurrence model. Therefore, leveraging the virus-mimicking nanoparticle to simulate the mechanism of virus-initiated specific immune responses to activate the intratumoral DCs may open up new strategies for in-situ cancer vaccines.
Collapse
Affiliation(s)
- Ya Chu
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Luyao Wang
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xi Huang
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Cenxi Lao
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Lin Luo
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jingyi Chen
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Fangfei Jin
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Lili Zhao
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Kaiyuan Hu
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Miao Lv
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xianjing Li
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yong Yang
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Weijun Zhao
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China; College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Wenguang Wang
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 210009, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
12
|
Tushe A, Marinelli E, Musca B, Ventura A, Zumerle S, Slukinova O, Zampardi G, Volpin F, Bonaudo C, Della Puppa A, Repellin M, Guerriero G, Lollo G, Mandruzzato S. Drug-loaded nanoparticles induce immunogenic cell death and efficiently target cells from glioblastoma patients. Nanomedicine (Lond) 2025:1-12. [PMID: 40326623 DOI: 10.1080/17435889.2025.2497747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
AIM Glioblastoma multiforme (GBM) is characterized by a highly immunosuppressive tumor microenvironment (TME), posing significant challenges for efficient therapy's outcomes. Nanomedicine combined with immunotherapy holds the potential to modulate the TME and reactivate immune responses. This study proposes a polymeric nanosystem (NPs) encapsulating diaminocyclohexane-platinum II (DACHPt), an oxaliplatin derivative, to induce immunogenic cell death (ICD) in GBM cells. MATERIALS & METHODS An ionic-gelation technique was employed to generate polymeric nanoparticles (NPs) with an approximate size of 200 nm. NPs internalization was analyzed in GBM cell lines, in vitro-derived macrophages, and in leukocytes and tumor cells from GBM patient via flow cytometry and confocal imaging. ICD was assessed by measuring two of its main markers: adenosine triphosphate (ATP) and high-mobility group box 1 (HMGB1). RESULTS NPs were efficiently incorporated by myeloid and tumor cells, but not by lymphocytes. DACHPt-loaded NPs demonstrated enhanced cytotoxicity compared to free drug, with increased ATP and HMGB1 release from GBM cells, confirming ICD induction. CONCLUSIONS Our findings suggest that DACHPt-loaded NPs represent a promising therapeutic strategy capable of targeting both tumor cells and tumor-promoting immune cells while inducing ICD.
Collapse
Affiliation(s)
- Ada Tushe
- Veneto Institute of Oncology IOV, IRCCS, Padua, Italy
| | | | | | | | - Sara Zumerle
- Veneto Institute of Oncology IOV, IRCCS, Padua, Italy
| | - Olga Slukinova
- Department of Surgery, Oncology and Gastroneterology, University of Padua, Padua, Italy
| | | | - Francesco Volpin
- Division of Neurosurgery, University Hospital of Padova, Padova, Italy
| | - Camilla Bonaudo
- Neurosurgery, Department of NEUROFARBA, University of Florence, University Hospital of Careggi, Florence, Italy
| | - Alessandro Della Puppa
- Neurosurgery, Department of NEUROFARBA, University of Florence, University Hospital of Careggi, Florence, Italy
| | - Mathieu Repellin
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, Lyon, France
| | - Giulia Guerriero
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, Lyon, France
| | - Giovanna Lollo
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, Lyon, France
- Institut universitaire de France (IUF), Paris, France
| | - Susanna Mandruzzato
- Veneto Institute of Oncology IOV, IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroneterology, University of Padua, Padua, Italy
| |
Collapse
|
13
|
Puri N, Sahane P, Phatale V, Khairnar P, Shukla S, Priyadarshinee A, Jain A, Srivastava S. Nano-chameleons: A review on cluster of differentiation-driven immune cell-engineered nanoarchitectonics for non-small cell lung cancer. Int J Biol Macromol 2025; 310:143440. [PMID: 40280523 DOI: 10.1016/j.ijbiomac.2025.143440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/26/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Cancer, being one of the most outrageous diseases, contributed to 48 % of the mortality in 2022, with lung cancer leading the race with a 12.4 % incidence rate. Conventional treatment modalities like radio-, chemo-, photo-, and immunotherapy employing nanocarriers often face several setbacks, such as non-specific delivery, off-site toxicity, rapid opsonization via the host immune system, and greater tumor recurrence rates. Moreover, the heterogeneous variability in the tumor microenvironment is responsible for existing therapy failure. With the advent of biomimetic nanoparticles as a novel and intriguing platform, researchers have exploited the inherent functionalities of the Cluster of Differentiation proteins (CD) as cell surface biomarkers and imparted the nanocarriers with enhanced homologous tumor targetability, immune evasion capability, and stealth properties, paving the way for improved therapy and diagnosis. This article explores pathogenesis and the multifaceted role of immune cells in non-small cell lung cancer. Moreover, the agenda of this article is to shed light on biomimetic nanoarchitectonics with respect to their fabrication, evaluation, and applications unraveling their synergistic effect with conventional therapies. Further discussion mentions the hurdles in clinical translation with viable solutions. The regulatory bottlenecks underscore the need for a regulatory roadmap with respect to commercialization. We believe that biomimetic nanoarchitectonics will be a beacon of hope in warfare against lung cancer.
Collapse
Affiliation(s)
- Niharika Puri
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Prajakta Sahane
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Shalini Shukla
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Abhipsa Priyadarshinee
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Akshita Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India.
| |
Collapse
|
14
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
15
|
Azmal M, Miah MM, Prima FS, Paul JK, Haque ASNB, Ghosh A. Advances and challenges in cancer immunotherapy: Strategies for personalized treatment. Semin Oncol 2025; 52:152345. [PMID: 40305928 DOI: 10.1016/j.seminoncol.2025.152345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 05/02/2025]
Abstract
Cancer immunotherapy has transformed oncology by harnessing the immune system to specifically target cancer cells, offering reduced systemic toxicity compared to traditional therapies. This review highlights key strategies, including adoptive cell transfer (ACT), immune checkpoint inhibitors, oncolytic viral (OV) therapy, monoclonal antibodies (mAbs), and mRNA-based vaccines. ACT reinfuses enhanced immune cells like tumor-infiltrating lymphocytes (TILs) to combat refractory cancers, while checkpoint inhibitors (eg, PD-1 and CTLA-4 blockers) restore T-cell activity. OV therapy uses engineered viruses (eg, T-VEC) to selectively lyse cancer cells, and advanced mAbs improve targeting precision. mRNA vaccines introduce tumor-specific antigens to trigger robust immune responses. Despite significant progress, challenges like immune-related side effects, high costs, and immunosuppressive tumor microenvironments persist. This review underscores the need for combination strategies and precision medicine to overcome these barriers and maximize the potential of immunotherapy in personalized cancer treatment.
Collapse
Affiliation(s)
- Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md Munna Miah
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Fatema Sultana Prima
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Anm Shah Newaz Been Haque
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh.
| |
Collapse
|
16
|
Zhu H, Chan CY, Heng JZX, Tang KY, Chai CHT, Tan HL, Loh XJ, Ye E, Li Z. Bioactive metal sulfide nanomaterials as photo-enhanced chemodynamic nanoreactors for tumor therapy. NANOSCALE HORIZONS 2025. [PMID: 40293306 DOI: 10.1039/d5nh00122f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Metal sulfide nanomaterials (MeSNs) are highly promising for biomedical applications due to their low toxicity, good dispersibility, high stability, adjustable particle sizes, and good biocompatibility. Their unique chemical and light-conversion properties also enable them to function as photothermal or photodynamic agents, enhancing chemodynamic therapy (CDT) of tumors. This makes MeSNs valuable as photo-enhanced CDT nanoagents, advancing precision and multi-modal tumor treatment. This review examines recent advancements in MeSNs for photo-enhanced chemodynamic tumor ablation, comparing their effectiveness in CDT. It highlights the roles of photothermal, photodynamic, and photocatalytic effects in enhancing treatment efficacy. MeSN-based nanoreactors are categorized by composition into iron sulfide, copper sulfide, other unary, and multi-MeSNs for their applications in tumor therapy. Additionally, this review discusses challenges, limitations, and future biomedical applications of MeSNs, offering insights into their potential for next-generation cancer treatments.
Collapse
Affiliation(s)
- Houjuan Zhu
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
| | - Chui Yu Chan
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
| | - Jerry Zhi Xiong Heng
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
| | - Karen Yuanting Tang
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
| | - Casandra Hui Teng Chai
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
| | - Hui Ling Tan
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2) A*STAR (Agency for Science, Technology and Research), Singapore 138634, Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
| | - Enyi Ye
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
| | - Zibiao Li
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, 138634, Singapore.
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2) A*STAR (Agency for Science, Technology and Research), Singapore 138634, Singapore
| |
Collapse
|
17
|
Rodella G, Préat V, Gallez B, Malfanti A. Design Strategies for Hyaluronic Acid-based Drug Delivery Systems in Cancer Immunotherapy. J Control Release 2025; 383:113784. [PMID: 40294800 DOI: 10.1016/j.jconrel.2025.113784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Despite its robust therapeutic potential, cancer immunotherapy has provided little progress towards improved survival rates for patients bearing immunologically refractory tumors. The implementation of advanced drug delivery systems represents a powerful means of improving cancer immunotherapy by relieving immunosuppression and promoting immune response; however, the overall impact of these systems on immunotherapy currently remains modest. Hyaluronic acid represents a widely used polymer in drug delivery; meanwhile, recent studies linking hyaluronic acid to the immune system make this polymer an attractive component in the design of next-generation cancer immunotherapies. Herein, we review our current understanding of the immunological properties of hyaluronic acid and discuss them in the context of bioactive functions and immune-related interactions with receptors, immune, and cancer cells. We analyze the potential of hyaluronic acid as a component in advanced drug delivery systems, highlighting strategies for the design of more effective vaccines and cancer chemo-immunotherapies. Finally, we discuss critical considerations to facilitate design and clinical translation to overcome existing challenges and maximize therapeutic potential.
Collapse
Affiliation(s)
- Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; Departement of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo, 5, 35131 Padova, Italy.
| |
Collapse
|
18
|
Adzavon KP, Zhao W, Khattak SN, Sheng W. Cholesterol-modified peptide nanomicelles as a promising platform for cancer therapy: A review. Int J Biol Macromol 2025; 311:143456. [PMID: 40274168 DOI: 10.1016/j.ijbiomac.2025.143456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/01/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Drug resistance, systemic toxicity, low solubility, and rapid clearance are common issues with chemotherapy drugs and other molecules used to treat cancer. The development of new therapeutic compounds and nanotherapy offers a solution to these issues. Therapeutic peptides have attracted great interest among these molecules due to their unique advantages, including low immunogenicity, efficient cellular internalization, deep tissue penetration, and low systemic toxicity. They have shown promise in cancer treatment by inducing apoptosis, necrosis, or cell lysis and promoting immunotherapy. In addition, peptides can deliver a range of cargoes, such as drugs, nucleic acids, imaging agents, and nanoparticles, and can specifically target cancer cells. However, problems such as their short half-life and low solubility limit their therapeutic use. Recent developments have addressed these constraints through structural alterations and nanoparticle formulations. In particular, cholesterol modification makes it possible for peptides to self-assemble into nanomicelles, which enhances their stability, half-life, and cell penetration. In this review, therapeutic peptides are presented as a versatile and successful cancer treatment option. The potential of cholesterol-modified peptide micelles as a reliable drug, nucleic acid, and imaging agent delivery system is also examined.
Collapse
Affiliation(s)
- Kodzo Prosper Adzavon
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Weijian Zhao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Sameena Noor Khattak
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Wang Sheng
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
19
|
Tong QS, Huang H, Yu HH, Liu R, Shen S, Du JZ. A size-switchable nanocluster remodels the immunosuppressive microenvironment of tumor and tumor-draining lymph nodes for improved cancer immunotherapy. Biomaterials 2025; 315:122910. [PMID: 39467399 DOI: 10.1016/j.biomaterials.2024.122910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Remodeling the immunosuppressive tumor microenvironment (TME) by immunomodulators has been well studied in the past years. However, strategies that enable concurrent modulation of both the immunosuppressive TME and tumor-draining lymph nodes (TDLNs) are still in the infancy. Here, we report a pH-sensitive size-switchable nanocluster, SPN-R848, to achieve simultaneous accumulation in tumor and TDLNs for immune activation. SPN-R848 with original size around 150 nm was formed by self-assembly of resiquimod (R848)-conjugated polyamidoamine (PAMAM) derivative, which could disintegrate into its small constituents (~ 8 nm) upon exposure to tumor acidity. The size reduction not only enhanced their accumulation and perfusion in the primary tumor, but promoted their transport and distribution in TDLNs. Accordingly, SPN-R848 remarkably remodeled the immunosuppressive TME by polarizing M2 to M1 macrophages and activated dendritic cells (DCs) in TDLNs, which synergistically facilitated the production and stimulation of cytotoxic T cells, and inhibited tumor growth in breast cancer and melanoma mouse models. Our study suggests that co-activation of immune microenvironments in both tumor and TDLNs may represent a promising direction to elicit strong antitumor immunity.
Collapse
Affiliation(s)
- Qi-Song Tong
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, 230031, China; School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Hua Huang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Hui-Han Yu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China.
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
20
|
Liu J, Liu J, Wang Y, Chen F, He Y, Xie X, Zhong Y, Yang C. Bioactive mesoporous silica materials-assisted cancer immunotherapy. Biomaterials 2025; 315:122919. [PMID: 39481339 DOI: 10.1016/j.biomaterials.2024.122919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/12/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Immunotherapy is initially envisioned as a powerful approach to train immune cells within the tumor microenvironment (TME) and lymphoid tissues to elicit strong anti-tumor responses. However, clinical cancer immunotherapy still faces challenges, such as limited immunogenicity and insufficient immune response. Leveraging the advantages of mesoporous silica (MS) materials in controllable drug and immunomodulator release, recent efforts have focused on engineering MS with intrinsic immunoregulatory functions to promote robust, systemic, and safe anti-tumor responses. This review discusses advances in bioactive MS materials that address the challenges of immunotherapy. Beyond their role in on-demand delivery and drug release in response to the TME, we highlight the intrinsic functions of bioactive MS in orchestrating localized immune responses by inducing immunogenic cell death in tumor cells, modulating immune cell activity, and facilitating tumor-immune cell interactions. Additionally, we emphasize the advantages of bioactive MS in recruiting and activating immune cells within lymphoid tissues to initiate anti-tumor vaccination. The review also covers the challenges of MS-assisted immunotherapy, potential solutions, and future outlooks. With a deeper understanding of material-bio interactions, the rational design of MS with sophisticated bioactivities and controllable responsiveness holds great promise for enhancing the outcomes of personalized immunotherapy.
Collapse
Affiliation(s)
- Jiali Liu
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Jiying Liu
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yaxin Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yan He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yiling Zhong
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Chao Yang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
21
|
Mamidi N, Franco De Silva F, Orash Mahmoudsalehi A. Advanced disease therapeutics using engineered living drug delivery systems. NANOSCALE 2025; 17:7673-7696. [PMID: 40040419 DOI: 10.1039/d4nr05298f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological barriers significantly impede the delivery of nanotherapeutics to diseased tissues, diminishing therapeutic efficacy across pathologies such as cancer and inflammatory disorders. Although conventional strategies integrate multifunctional designs and molecular components into nanomaterials (NMs), many approaches remain insufficient to overcome these barriers. Key challenges, including inadequate drug accumulation at target sites and nonspecific biodistribution, persist in nanotherapeutic development. NMs, which harness the ability to precisely modulate drug delivery spatiotemporally and control release kinetics, represent a transformative platform for targeted cancer therapy. In this review, we highlight the biological obstacles limiting effective cancer treatment and evaluate how stimuli-responsive NMs address these constraints. By leveraging exogenous and endogenous stimuli, such NMs improve therapeutic specificity, reduce off-target effects, and amplify drug activity within pathological microenvironments. We systematically analyze the rational design and synthesis of stimuli-responsive NMs, driven by advances in oncology, biomaterials science, and nanoscale engineering. Furthermore, we highlight advances across NM classes-including polymeric, lipid-based, inorganic, and hybrid systems and explore functionalization approaches using targeting ligands, antibodies, and biomimetic coatings. Diverse delivery strategies are evaluated, such as small-molecule prodrug activation, peptide- and protein-based targeting, nucleic acid payloads, and engineered cell-mediated transport. Despite the promise of stimuli-responsive NMs, challenges such as biocompatibility, scalable fabrication, and clinical translation barriers must be addressed. By elucidating structure-function relationships and refining stimulus-triggered mechanisms, these NMs pave the way for transformative precision oncology strategies, enabling patient-specific therapies with enhanced efficacy and safety. This synthesis of interdisciplinary insights aims to catalyze innovation in next-generation nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for Nanobiosystems, School of Pharmacy, University of Wisconsin-Madison, Wisconsin-53705, USA.
| | - Fátima Franco De Silva
- Department of Food Engineering, Tecnologico de Monterrey, Monterrey, Nuevo Leon-64849, Mexico
| | - Amin Orash Mahmoudsalehi
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo Leon-64849, Mexico
| |
Collapse
|
22
|
Steger K, Fiegl H, Feroz B, Leitner K, Marth C, Hackl H, Zeimet AG. Differences in immunogenicity of TP53-mutated cancers with low tumor mutational burden (TMB) A study on TP53mut endometrial-, ovarian- and triple-negative breast cancer. Eur J Cancer 2025; 219:115320. [PMID: 39986022 DOI: 10.1016/j.ejca.2025.115320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
PURPOSE To explore why in large phase III randomized clinical trials TP53-mutated (TP53mut) endometrial cancer (EC) was the only tumor showing survival benefit to immune checkpoint inhibitors (ICIs) added to chemotherapy when compared with other low TMB TP53mut cancers, such as high-grade serous ovarian (HGSOC) and triple-negative breast cancer (TNBC). EXPERIMENTAL DESIGN From 606 patients with one of the three mentioned cancers, "The Cancer Genome Atlas" data on clinical outcome, TMB and detailed composition of the tumor immune-microenvironment (TIME) (immune infiltrating cells, cytokines, and other immune-modulators) were compared using the Kruskal-Wallis test, followed by Pearson correlation. Prognostic value of studied variables was assessed by Kaplan-Meier and Cox-regression analyses. RESULTS TMB was very low in all three TP53mut entities, being lowest in EC (median: 1.27 Mut/Mb; p < 0.001). Interestingly, high TMB was significantly associated with improved clinical outcome in every entity, whereby best discrimination for PFS was found in EC (HR: 0.52). Compared to EC, immune-suppressing regulatory T-cells were higher in HGSOC and TNBC (p < 0.001) and M2-like macrophages higher in HGSOC (p < 0.001). In contrast, immune-activating mDCs were more prominent in EC than in HGSOC (p < 0.001). Differential modulator expression analyses revealed highest discrimination for the immune-inhibiting FOXP3, C1QA and XBP1, which all exhibited lower levels in EC compared with HGSOC and TNBC (p < 0.001). CONCLUSION Characteristics of TIME differ substantially among the assessed entities in terms that EC exhibits fewer immunosuppressive traits, expecting a higher likelihood for responding to ICIs, despite a very low TMB, whereas HGSOC and TNBC exhibit an immune hostile TIME.
Collapse
Affiliation(s)
- Katharina Steger
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Heidelinde Fiegl
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barin Feroz
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Leitner
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Marth
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Hackl
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Alain G Zeimet
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
23
|
Bayat M, Nahand JS. Battlegrounds of treatment resistance: decoding the tumor microenvironment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04055-5. [PMID: 40131387 DOI: 10.1007/s00210-025-04055-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
The tumor microenvironment (TME) emerges as a formidable actor in the cancer treatment landscape, wielding the power to thwart therapeutic efficacy across various modalities, including chemotherapy, radiotherapy, immunotherapy, targeted therapy, and hormonal therapy. This intricate ecosystem comprising diverse cellular constituents, signaling molecules, and the extracellular matrix fosters a dynamic interplay that profoundly influences tumor behavior and treatment outcomes. This review explores the mechanisms through which the TME drives resistance to standard therapies, emphasizing key factors such as hypoxia, immune evasion, and metabolic reprogramming. Furthermore, we illuminate innovative strategies aimed at reprogramming this hostile environment, including the application of therapeutic vaccines, CAR T cell therapy, and combination immunotherapies designed to enhance anti-tumor responses. By advocating for multidimensional approaches that dismantle the TME's barriers to effective treatment, this review calls for a transformative shift in cancer treatment paradigms. By bridging the gap between the TME's complexities and targeted therapeutic strategies, we pave the way for targeted interventions that promise to enhance clinical outcomes and improve patient prognosis in the relentless battle against cancer.
Collapse
Affiliation(s)
- Mobina Bayat
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Shah DD, Chorawala MR, Raghani NR, Patel R, Fareed M, Kashid VA, Prajapati BG. Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med Oncol 2025; 42:117. [PMID: 40102282 DOI: 10.1007/s12032-025-02641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Tumor microenvironment (TME) denotes the non-cancerous cells and components presented in the tumor, including molecules produced and released by them. Interactions between cancer cells, immune cells, stromal cells, and the extracellular matrix within the TME create a dynamic ecosystem that can either promote or hinder tumor growth and spread. The TME plays a pivotal role in either promoting or inhibiting tumor growth and dissemination, making it a critical factor to consider in the development of effective cancer therapies. Understanding the intricate interplay within the TME is crucial for devising effective cancer therapies. Combination therapies involving inhibitors of immune checkpoint blockade (ICB), and/or chemotherapy now offer new approaches for cancer therapy. However, it remains uncertain how to best utilize these strategies in the context of the complex tumor microenvironment. Oncogene-driven changes in tumor cell metabolism can impact the TME to limit immune responses and present barriers to cancer therapy. Cellular and acellular components in tumor microenvironment can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Components in the TME can reprogram tumor behavior and influence responses to treatments, facilitating immune evasion, nutrient deprivation, and therapeutic resistance. Moreover, the TME can influence angiogenesis, promoting the formation of blood vessels that sustain tumor growth. Notably, the TME facilitates immune evasion, establishes a nutrient-deprived milieu, and induces therapeutic resistance, hindering treatment efficacy. A paradigm shift from a cancer-centric model to a TME-centric one has revolutionized cancer research and treatment. However, effectively targeting specific cells or pathways within the TME remains a challenge, as the complexity of the TME poses hurdles in designing precise and effective therapies. This review highlights challenges in targeting the tumor microenvironment to achieve therapeutic efficacy; explore new approaches and technologies to better decipher the tumor microenvironment; and discuss strategies to intervene in the tumor microenvironment and maximize therapeutic benefits.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Rajanikant Patel
- Department of Product Development, Granules Pharmaceuticals Inc., 3701 Concorde Parkway, Chantilly, VA, 20151, USA
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 13713, Riyadh, Saudi Arabia
| | - Vivekanand A Kashid
- MABD Institute of Pharmaceutical Education and Research, Babhulgaon, Yeola, Nashik, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
25
|
Yates J, Mathey-Andrews C, Park J, Garza A, Gagné A, Hoffman S, Bi K, Titchen B, Hennessey C, Remland J, Shannon E, Camp S, Balamurali S, Cavale SK, Li Z, Raghawan AK, Kraft A, Boland G, Aguirre AJ, Sethi NS, Boeva V, Van Allen E. Cell states and neighborhoods in distinct clinical stages of primary and metastatic esophageal adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.17.608386. [PMID: 39229240 PMCID: PMC11370330 DOI: 10.1101/2024.08.17.608386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Esophageal adenocarcinoma (EAC) is a highly lethal cancer of the upper gastrointestinal tract with rising incidence in western populations. To decipher EAC disease progression and therapeutic response, we performed multiomic analyses of a cohort of primary and metastatic EAC tumors, incorporating single-nuclei transcriptomic and chromatin accessibility sequencing, along with spatial profiling. We identified tumor microenvironmental features previously described to associate with therapy response. We identified five malignant cell programs, including undifferentiated, intermediate, differentiated, epithelial-to-mesenchymal transition, and cycling programs, which were associated with differential epigenetic plasticity and clinical outcomes, and for which we inferred candidate transcription factor regulons. Furthermore, we revealed diverse spatial localizations of malignant cells expressing their associated transcriptional programs and predicted their significant interactions with microenvironmental cell types. We validated our findings in three external single-cell RNA-seq and three bulk RNA-seq studies. Altogether, our findings advance the understanding of EAC heterogeneity, disease progression, and therapeutic response.
Collapse
Affiliation(s)
- Josephine Yates
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Camille Mathey-Andrews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Amanda Garza
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Andréanne Gagné
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Samantha Hoffman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Breanna Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
| | | | - Joshua Remland
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Sabrina Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Siddhi Balamurali
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shweta Kiran Cavale
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Zhixin Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Akhouri Kishore Raghawan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Agnieszka Kraft
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Genevieve Boland
- Department of Surgery, Division of Gastrointestinal and Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Nilay S Sethi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Valentina Boeva
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
- Cochin Institute, Inserm U1016, CNRS UMR 8104, Paris Descartes University UMR-S1016, Paris 75014, France
| | - Eliezer Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Gao M, Sun Q, Zhang R, Shan G, Zhang H, Peng R, Liu M, Sun G, Qiao L, Li Y, He X. Extracellular vesicles-hitchhiking boosts the deep penetration of drugs to amplify anti-tumor efficacy. Biomaterials 2025; 314:122829. [PMID: 39276410 DOI: 10.1016/j.biomaterials.2024.122829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Developing drug delivery systems capable of achieving deep tumor penetration is a challenging task, yet there is a significant demand for such systems in cancer treatment. Hitchhiking on tumor-derived extracellular vesicles (EVs) represents a promising strategy for enhancing drug penetration into tumors. However, the limited drug assembly on EVs restricts its further application. Here, we present a novel approach to efficiently attach antitumor drugs to EVs using an engineered cell membrane-based vector. This vector includes the AS1411 aptamer for tumor-specific targeting, the vesicular stomatitis virus glycoprotein (VSV-G) for tumor cell membrane fusion, and a photosensitizer as the therapeutic agent while ensuring optimal drug encapsulation and stability. Upon injection, photosensitizers are firstly transferred to the tumor cell membrane and subsequently piggybacked onto EVs with the inherent secretion process. By hitchhiking with EVs, photosensitizers can be transferred layer by layer deep into the solid tumors. The results suggest that this EVs-hitchhiking strategy enables photosensitizers to penetrate deeply into tumor tissue, thereby enhancing the efficacy of phototherapy. This study offers broad application prospects for delivering drugs deeply into tumor tissues.
Collapse
Affiliation(s)
- Min Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Qiuting Sun
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Ruijie Zhang
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Guisong Shan
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Huiru Zhang
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Rui Peng
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Mengyu Liu
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Lei Qiao
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| | - Yang Li
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China.
| | - Xiaoyan He
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China.
| |
Collapse
|
27
|
Luo Y, Linghu M, Luo X, Li D, Wang J, Peng S, Ma Y. Remodeling tumor immunosuppressive microenvironment through dual activation of immunogenic panoptosis and ferroptosis by H 2S-amplified nanoformulation to enhance cancer immunotherapy. Acta Pharm Sin B 2025; 15:1242-1254. [PMID: 40370544 PMCID: PMC12069112 DOI: 10.1016/j.apsb.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 05/16/2025] Open
Abstract
The deficiency in immunogenicity and the presence of immunosuppression within the tumor microenvironment significantly hindered the efficacy of immunotherapy. Consequently, a nanoformulation containing metal sulfide of FeS and GSDMD plasmid (NPFeS/GD) had been developed to effectively augment antitumor immune responses through dual activation of immunogenic PANoptosis and ferroptosis, as well as reprogramming immunosuppressive effects via H2S amplification. The bioactive NPFeS/GD exhibited controlled release of GSDMD plasmid, H2S, and Fe2+ in response to the tumor microenvironment. Fe2+, H2S, and the expression of GSDMD protein could effectively elicit highly immunogenic PANoptosis and ferroptosis. Furthermore, releasing H2S could mitigate the overexpression of indoleamine 2,3-dioxygenase1 (IDO1) induced by immunogenic PANoptotic and ferroptotic cell death and disrupt the activity of IDO1. Consequently, NPFeS/GD effectively triggered the antitumor innate and adaptive immune responses through induction of PANoptotic and ferroptotic cell death and reshaped the tumor immunosuppressive microenvironment to enhance antitumor immunotherapy for metastasis inhibition. This study unveiled the significant potential of immunogenic PANoptosis and ferroptosis in H2S gas therapy for enhancing tumor immunotherapy, offering novel insights and ideas for the rational design of nanomedicine to enhance tumor immunogenicity while reprogramming the tumor immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Yingli Luo
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi 214062, China
| | - Maoyuan Linghu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xianyu Luo
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Dongdong Li
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Shaojun Peng
- Center for Biological Science and Technology & College of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
| | - Yinchu Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi 214062, China
| |
Collapse
|
28
|
D'Amico C, Fusciello M, Hamdan F, D'Alessio F, Bottega P, Saklauskaite M, Russo S, Cerioni J, Elbadri K, Kemell M, Hirvonen J, Cerullo V, Santos HA. Transdermal delivery of PeptiCRAd cancer vaccine using microneedle patches. Bioact Mater 2025; 45:115-127. [PMID: 39639878 PMCID: PMC11617629 DOI: 10.1016/j.bioactmat.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
Microneedles (MNs) are a prospective system in cancer immunotherapy to overcome barriers regarding proper antigen delivery and presentation. This study aims at identifying the potential of MNs for the delivery of Peptide-coated Conditionally Replicating Adenoviruses (PeptiCRAd), whereby peptides enhance the immunogenic properties of adenoviruses presenting tumor associated antigens. The combination of PeptiCRAd with MNs containing polyvinylpyrrolidone and sucrose was tested for the preservation of structure, induction of immune response, and tumor eradication. The findings indicated that MN-delivered PeptiCRAd was effective in peptide presentation in vivo, leading to complete tumor rejection when mice were pre-vaccinated. A rise in the cDC1 population in the lymph nodes of the MN treated mice led to an increase in the effector memory T cells in the body. Thus, the results of this study demonstrate that the combination of MN technology with PeptiCRAd may provide a safer, more tolerable, and efficient approach to cancer immunotherapy, potentially translatable to other therapeutic applications.
Collapse
Affiliation(s)
- Carmine D'Amico
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Manlio Fusciello
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
| | - Firas Hamdan
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
| | - Federica D'Alessio
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
| | - Paolo Bottega
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
| | - Milda Saklauskaite
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
| | - Salvatore Russo
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
| | - Justin Cerioni
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
| | - Khalil Elbadri
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Marianna Kemell
- Department of Chemistry, Faculty of Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Vincenzo Cerullo
- Department of Pharmaceutical Biosciences, University of Helsinki, Faculty of Pharmacy ImmunoViroTherapy Lab, Drug Research Program, Viikinkaari 5, E00790, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Fabianinkatu 33, 00710, Helsinki, Finland
- Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, 00014, Helsin-ki, Finland
- Department of Molecular Medicine and Medical Biotechnology and CEINGE, Naples University Federico II, 80131, Naples, Italy
| | - Hélder A. Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV, Groningen, the Netherlands
| |
Collapse
|
29
|
Luo X, Zhang Y, Zeng Y, Yang D, Zhou Z, Zheng Z, Xiao P, Ding X, Li Q, Chen J, Deng Q, Zhong X, Qiu S, Yan W. Nanotherapies Based on ROS Regulation in Oral Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409087. [PMID: 39887942 PMCID: PMC11884622 DOI: 10.1002/advs.202409087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/28/2024] [Indexed: 02/01/2025]
Abstract
Oral diseases rank among the most prevalent clinical conditions globally, typically involving detrimental factors such as infection, inflammation, and injury in their occurrence, development, and outcomes. The concentration of reactive oxygen species (ROS) within cells has been demonstrated as a pivotal player in modulating these intricate pathological processes, exerting significant roles in restoring oral functionality and maintaining tissue structural integrity. Due to their enzyme-like catalytic properties, unique composition, and intelligent design, ROS-based nanomaterials have garnered considerable attention in oral nanomedicine. Such nanomaterials have the capacity to influence the spatiotemporal dynamics of ROS within biological systems, guiding the evolution of intra-ROS to facilitate therapeutic interventions. This paper reviews the latest advancements in the design, functional customization, and oral medical applications of ROS-based nanomaterials. Through the analysis of the components and designs of various novel nanozymes and ROS-based nanoplatforms responsive to different stimuli dimensions, it elaborates on their impacts on the dynamic behavior of intra-ROS and their potential regulatory mechanisms within the body. Furthermore, it discusses the prospects and strategies of nanotherapies based on ROS scavenging and generation in oral diseases, offering alternative insights for the design and development of nanomaterials for treating ROS-related conditions.
Collapse
Affiliation(s)
- Xin Luo
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yanli Zhang
- Stomatological HospitalSchool of StomatologySouthern Medical UniversityGuangzhou510280China
| | - Yuting Zeng
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Dehong Yang
- Department of Orthopedics Spinal SurgeryNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Zhiyan Zhou
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Ziting Zheng
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Ping Xiao
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Xian Ding
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Qianlin Li
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Jiaping Chen
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Qianwen Deng
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Xincen Zhong
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Sijie Qiu
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Wenjuan Yan
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
30
|
Parra-Nieto J, Hidalgo L, Márquez-Cantudo M, García-Castro J, Megias D, Ramirez M, Baeza A. Liposomal-Based Nanoarchitectonics as Bispecific T Cell Engagers in Neuroblastoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11937-11945. [PMID: 39957209 DOI: 10.1021/acsami.5c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
Neuroblastoma (NB) is an aggressive pediatric solid tumor that lacks efficient treatment. In the past few years, the use of engineered lymphocytes endowed with chimeric antigen receptors (CAR T), which improve their natural search and destroy skills against tumoral cells, has provided a highly valuable strategy to eradicate tumors in a specific and safe manner. Unfortunately, despite the excellent results achieved by these cell-based therapies in liquid tumors, their efficacy in the treatment of solid malignancies is usually modest due to the existence of several biological barriers which compromise their efficacy. Herein, a strategy to guide CAR T toward NB cells based on the use of nanometric bispecific T engagers (NBTEs) is presented. These novel bispecific nanoplatforms are based on liposomes and protocells doubly functionalized with synthetic targeting moieties (para-aminobenzylguanidine and fluorescein) able to selectively bind to membrane cell receptors of NB and anti-FITC CAR T, respectively. The binding process of NBTEs to NB cells was monitored by confocal fluorescence microscopy showing the excellent capacity of these nanodevices to place fluorescence labels on the surface of the malignant cells. Then, NB cells previously incubated in the presence of NBTEs were rapidly detected and destroyed by anti-FITC CAR T, which confirmed the excellent capacity of these nanoplatforms to improve the natural capacity of CAR T to eradicate malignant cells. Finally, the high versatility of the NBTE design and its easy-to-tune nature would allow their rapid application to different types of solid tumors.
Collapse
Affiliation(s)
- Jorge Parra-Nieto
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Laura Hidalgo
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Marta Márquez-Cantudo
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Javier García-Castro
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Diego Megias
- Advanced Optical Microscopy Unit, Instituto de salud Carlos III (ISCIII), Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Manuel Ramirez
- Servicio de Hematología y Oncología Pediátrica, Hospital Infantil Universitario Niño Jesús, Av. de Menéndez Pelayo, 65, Retiro, 28009 Madrid, Spain
| | - Alejandro Baeza
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
31
|
Luo J, Yi T, Wang Y, Song W, Gao Z, Wang J, Li Y. ESM1 promote proliferation, invasion and angiogenesis via Akt/mTOR and Ras pathway in kidney renal clear cell carcinoma. Sci Rep 2025; 15:4902. [PMID: 39929852 PMCID: PMC11811180 DOI: 10.1038/s41598-024-82400-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 12/05/2024] [Indexed: 02/13/2025] Open
Abstract
The most common types of renal carcinoma is kidney renal clear cell carcinoma (KIRC). ESM1 is a secreted protein, which involved in, lipids and glucose metabolism, but their role in angiogenesis is contradictory in different disease, especially in KIRC. Bioinformatic analysis confirmed the ESM1 expression and prognosis in KIRC. IHC staining revealed protein expression of ESM1 in KIRC samples. The role of ESM1 in KIRC proliferation and migration were tested by MTT, EdU, transwell analysis. The role of its paracrine function in KIRC angiogenesis was tested by functional experiments. The downstream molecular mechanism of ESM1 were further elucidated by WB and functional experiments. ESM1 was significantly increased in KIRC with prognostic significance. ESM1 knockdown inhibited the invasiveness capability and viability of KIRC cell. The paracrine of ESM1 enhanced HUVECs proliferation and migration to format tube in KIRC cell conditional medium. ESM1 knockdown induced the inactivation of Akt/mTOR and Ras pathway to attenuate proliferation, migration, invasion and angiogenesis in KIRC. ESM1 was a key role in the tumor microenvironment (TME) of KIRC, which promoted the proliferation, migration, invasion, and angiogenesis by activating Akt/mTOR and Ras pathway. It is a potential therapeutic target for KIRC patients.
Collapse
Affiliation(s)
- Jianjun Luo
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan Province, China
| | - Ting Yi
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Department of Trauma Center, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Yong Wang
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan Province, China
| | - Wei Song
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan Province, China
| | - Zhiyong Gao
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan Province, China
| | - Jiansong Wang
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan Province, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
32
|
Agrawal A, Javanmardi Y, Watson SA, Serwinski B, Djordjevic B, Li W, Aref AR, Jenkins RW, Moeendarbary E. Mechanical signatures in cancer metastasis. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:3. [PMID: 39917412 PMCID: PMC11794153 DOI: 10.1038/s44341-024-00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/20/2024] [Indexed: 02/09/2025]
Abstract
The cancer metastatic cascade includes a series of mechanical barrier-crossing events, involving the physical movement of cancer cells from their primary location to a distant organ. This review describes the physical changes that influence tumour proliferation, progression, and metastasis. We identify potential mechanical signatures at every step of the metastatic cascade and discuss some latest mechanobiology-based therapeutic interventions to highlight the importance of interdisciplinary approaches in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ayushi Agrawal
- Department of Mechanical Engineering, University College London, London, UK
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, UK
| | - Sara A. Watson
- Department of Mechanical Engineering, University College London, London, UK
- Division of Biosciences, University College London, London, UK
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, UK
- Northeastern University London, London, UK
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, UK
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Amir R. Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Russell W. Jenkins
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| |
Collapse
|
33
|
Liu T, Lu C, Jiang X, Wang Y, Chen Z, Qi C, Xu X, Feng X, Wang Q. Nano-Based Strategies Aiming at Tumor Microenvironment for Improved Cancer Therapy. Mol Pharm 2025; 22:647-677. [PMID: 39818981 DOI: 10.1021/acs.molpharmaceut.4c01267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Malignant tumors pose a considerable threat to human life and health. Traditional treatments, such as radiotherapy and chemotherapy, often lack specificity, leading to collateral damage to normal tissues. Tumor microenvironment (TME) is characterized by hypoxia, acidity, redox imbalances, and elevated ATP levels factors that collectively promote tumor growth and metastasis. This review provides a comprehensive overview of the nanoparticles developed in recent years for TME-responsive strategies or TME-modulating methods for tumor therapy. The TME-responsive strategies focus on designing and synthesizing nanoparticles that can interact with the tumor microenvironment to achieve precisely controlled drug release. These nanoparticles activate drug release under specific conditions within the tumor environment, thereby enhancing the efficacy of the drugs while reducing toxicity to normal cells. Moreover, simply eliminating tumor cells does not fundamentally solve the problem. Only by comprehensively regulating the TME to make it unsuitable for tumor cell survival and proliferation can we achieve more thorough therapeutic effects and reduce the risk of tumor recurrence. TME regulation strategies aim to suppress the growth and metastasis of tumor cells by modulating various components within the TME. These strategies not only improve treatment outcomes but also have the potential to lay the foundation for future personalized cancer therapies.
Collapse
Affiliation(s)
- Tianhui Liu
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Changshun Lu
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Xue Jiang
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Yutong Wang
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Zhengrong Chen
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Chunshuang Qi
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Xiaoru Xu
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun 130117, China
| | - Xiangru Feng
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Qingshuang Wang
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| |
Collapse
|
34
|
Zhong W, Zhang G, Yue K, Song Y, Zhao Z. MMP2 enzyme-responsive extracellular vesicles as dual-targeted carriers to promote the phagocytosis of macrophages. Colloids Surf B Biointerfaces 2025; 246:114365. [PMID: 39531838 DOI: 10.1016/j.colsurfb.2024.114365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Combination therapy using inhibition of tumor cell escape and alteration of the tumor microenvironment offers a new strategy for cancer treatment. This study aimed to develop an extracellular vesicle (EV) carrier that regulates tumor cells and the tumor microenvironment to achieve efficient tumor immunotherapy. The ligand modified on carriers targets the immune checkpoint CD47 protein, blocking tumor cell escape. This ligand is cleaved by the MMP2 enzyme and assembles into nanofibers, extending the retention time in the tumor. The carriers target the CD206 protein, enabling efficient uptake by M2 macrophages. Carriers with a high density of ligands (anti-CD206) exhibit strong receptorligand interactions with tumor cells. Due to their high rigidity, these EVs have difficulty deforming during the transmembrane process, reducing resistance and resulting in low uptake efficiency by M2 cells. The optimal uptake efficiency by M2 macrophages is achieved when the mass ratio of ligand to EVs is 1:25. Crocin loaded in EVs facilitates the polarization of M2 macrophages into M1 cells, which can phagocytize tumor cells. This study reveals a potential strategy for using extracellular vesicles in tumor treatment.
Collapse
Affiliation(s)
- Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong 528399, China.
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center, China; National Clinical Research Center for Cancer, China; Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center, China; National Clinical Research Center for Cancer, China; Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
35
|
Jawaid S, Joshi Y, Neelofar N, Khursheed K, Shams S, Chaudhary M, Arora M, Mahajan K, Anwar F. A Cross-talk between Nanomedicines and Cardiac Complications: Comprehensive View. Curr Pharm Des 2025; 31:741-752. [DOI: https:/doi.org/10.2174/0113816128347223241021111914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/24/2024] [Indexed: 05/15/2025]
Abstract
Background:
Cardiovascular Diseases (CVDs) are the leading cause of global morbidity and mortality,
necessitating innovative approaches for both therapeutics and diagnostics. Nanoscience has emerged as a
promising frontier in addressing the complexities of CVDs.
Objective:
This study aims to explorethe interaction of CVDs and Nanomedicine (NMs), focusing on applications
in therapeutics and diagnostics.
Observations:
In the realm of therapeutics, nanosized drug delivery systems exhibit unique advantages, such
as enhanced drug bioavailability, targeted delivery, and controlled release. NMs platform, including liposomes,
nanoparticles, and carriers, allows the precise drug targeting to the affected cardiovascular tissues with
minimum adverse effects and maximum therapeutic efficacy. Moreover, nanomaterial (NM) enables the integration
of multifunctional components, such as therapeutic agents and target ligands, into a single system for
comprehensive CVD management. Diagnostic fronts of NMs offer innovative solutions for early detection and
monitoring of CVDs. Nanoparticles and nanosensors enable highly sensitive and specific detection of Cardiac
biomarkers, providing valuable insights into a disease state, its progression, therapeutic outputs, etc. Further,
nano-based technology via imaging modalities offers high high-resolution imaging, aiding in the vascularization
of cardiovascular structures and abnormalities. Nanotechnology-based imaging modalities offer high-resolution
imaging and aid in the visualization of cardiovascular structures and abnormalities.
Conclusion:
The cross-talk of CVDs and NMs holds tremendous potential for revolutionizing cardiovascular
healthcare by providing targeted and efficient therapeutic interventions, as well as sensitive and early detection
for the improvement of patient health if integrated with Artificial Intelligence (AI).
Collapse
Affiliation(s)
- Shagufta Jawaid
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Yogesh Joshi
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Nauroz Neelofar
- Department of Obstetrics and Gynae, Himaliyan Institute of Medical Sciences, Swami Rama Himaliyan University, Jollygrand,
Dehradun, Uttarakhand, India
| | - Khuzamah Khursheed
- Shri Guru Ram Rai Institute of Medical and Health Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand, India
| | - Samya Shams
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Mansi Chaudhary
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Mitali Arora
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Karan Mahajan
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah,
Saudi Arabia
| |
Collapse
|
36
|
Jang I, Yum K, Han S, Moon S, Lee JB. A virus-inspired RNA mimicry approach for effective cancer immunotherapy. J Mater Chem B 2025; 13:1619-1629. [PMID: 39834198 DOI: 10.1039/d4tb02301c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Current cancer treatments, including chemotherapy, surgery, and radiation, often present significant challenges such as severe side effects, drug resistance, and damage to healthy tissues. To address these issues, we introduce a virus-inspired RNA mimicry approach, specifically through the development of uridine-rich nanoparticles (UNPs) synthesized using the rolling circle transcription (RCT) technique. These UNPs are designed to mimic the poly-U tail sequences of viral RNA, effectively engaging RIG-I-like receptors (RLRs) such as MDA5 and LGP2 in cancer cells. Activation of these receptors leads to the upregulation of pro-inflammatory cytokines and the initiation of apoptosis, resulting in targeted cancer cell death. Importantly, this strategy overcomes the limitations of traditional therapies and enhances the effectiveness of existing RIG-I stimulators, such as poly(I:C), which has often exhibited toxicity in clinical settings due to delivery methods. Our in vivo studies further demonstrate the ability of UNPs to significantly reduce tumor growth without adverse effects, highlighting their potential as a novel and effective approach in cancer immunotherapy. This approach offers new therapeutic strategies that leverage the body's innate antiviral mechanisms for cancer treatment.
Collapse
Affiliation(s)
- Iksoo Jang
- Department of Chemical Engineering, University of Seoul, Republic of Korea
| | - Kyuha Yum
- Department of Chemical Engineering, University of Seoul, Republic of Korea
| | - Sangwoo Han
- Department of Chemical Engineering, University of Seoul, Republic of Korea
| | - Sunghyun Moon
- Department of Chemical Engineering, University of Seoul, Republic of Korea
| | - Jong Bum Lee
- Department of Chemical Engineering, University of Seoul, Republic of Korea
- Center for Innovative Chemical Processes, Institute of Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea.
| |
Collapse
|
37
|
Farooq MA, Johnston APR, Trevaskis NL. Impact of nanoparticle properties on immune cell interactions in the lymph node. Acta Biomater 2025; 193:65-82. [PMID: 39701340 DOI: 10.1016/j.actbio.2024.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
The lymphatic system plays an important role in health and many diseases, such as cancer, autoimmune, cardiovascular, metabolic, hepatic, viral, and other infectious diseases. The lymphatic system is, therefore, an important treatment target site for a range of diseases. Lymph nodes (LNs), rich in T cells, B cells, dendritic cells, and macrophages, are also primary sites of action for vaccines and immunotherapies. Promoting the delivery of therapeutics and vaccines to LNs can, therefore, enhance treatment efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. Several nanoparticle (NP) based delivery systems, such as polymeric NPs, lipid NPs, liposomes, micelles, and dendrimers, have been reported to enhance the delivery of therapeutics and/or vaccines to LNs. Specific uptake into the lymph following injection into tissues is highly dependent on particle properties, particularly particle size, as small molecules are more likely to be taken up by blood capillaries due to higher blood flow rates, whereas larger molecules and NPs can be specifically transported via the lymphatic vessels to LNs as the initial lymphatic capillaries are more permeable than blood capillaries. Once NPs enter LNs, particle properties also have an important influence on their disposition within the node and association with immune cells, which has significant implications for the design of vaccines and immunotherapies. This review article focuses on the impact of NP properties, such as size, surface charge and modification, and route of administration, on lymphatic uptake, retention, and interactions with immune cells in LNs. We suggest that optimizing all these factors can enhance the efficacy of vaccines or therapeutics with targets in the lymphatics and also be helpful for the rational design of vaccines. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an essential role in health and is an important treatment target site for a range of diseases. Promoting the delivery of immunotherapies and vaccines to immune cells in lymph nodes can enhance efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. One of the major approaches used to deliver therapeutics and vaccines to lymph nodes is via injection in nanoparticle delivery systems. This review aims to provide an overview of the impact of nanoparticle properties, such as size, surface charge, modification, and route of administration, on lymphatic uptake, lymph node retention, and interactions with immune cells in lymph nodes. This will inform the design of future improved nanoparticle systems for vaccines and immunotherapies.
Collapse
Affiliation(s)
- Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
38
|
Ma M, Zhang Y, Pu K, Tang W. Nanomaterial-enabled metabolic reprogramming strategies for boosting antitumor immunity. Chem Soc Rev 2025; 54:653-714. [PMID: 39620588 DOI: 10.1039/d4cs00679h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Immunotherapy has become a crucial strategy in cancer treatment, but its effectiveness is often constrained. Most cancer immunotherapies focus on stimulating T-cell-mediated immunity by driving the cancer-immunity cycle, which includes tumor antigen release, antigen presentation, T cell activation, infiltration, and tumor cell killing. However, metabolism reprogramming in the tumor microenvironment (TME) supports the viability of cancer cells and inhibits the function of immune cells within this cycle, presenting clinical challenges. The distinct metabolic needs of tumor cells and immune cells require precise and selective metabolic interventions to maximize therapeutic outcomes while minimizing adverse effects. Recent advances in nanotherapeutics offer a promising approach to target tumor metabolism reprogramming and enhance the cancer-immunity cycle through tailored metabolic modulation. In this review, we explore cutting-edge nanomaterial strategies for modulating tumor metabolism to improve therapeutic outcomes. We review the design principles of nanoplatforms for immunometabolic modulation, key metabolic pathways and their regulation, recent advances in targeting these pathways for the cancer-immunity cycle enhancement, and future prospects for next-generation metabolic nanomodulators in cancer immunotherapy. We expect that emerging immunometabolic modulatory nanotechnology will establish a new frontier in cancer immunotherapy in the near future.
Collapse
Affiliation(s)
- Muye Ma
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Dr 2, Singapore, 117545, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Dr, Singapore, 117597, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Wei Tang
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
- Department of Pharmacy and Pharmaceutic Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| |
Collapse
|
39
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
40
|
Shao X, Zhao X, Wang B, Fan J, Wang J, An H. Tumor microenvironment targeted nano-drug delivery systems for multidrug resistant tumor therapy. Theranostics 2025; 15:1689-1714. [PMID: 39897552 PMCID: PMC11780529 DOI: 10.7150/thno.103636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
In recent years, nano-drug delivery systems (Nano-DDS) that target the tumor microenvironment (TME) to overcome multidrug resistance (MDR) have become a research hotspot in the field of cancer therapy. By precisely targeting the TME and regulating its unique pathological features, such as hypoxia, weakly acidic pH, and abnormally expressed proteins, etc., these Nano-DDS enable effective delivery of therapeutic agents and reversal of MDR. This scientific research community is increasing its investment in the development of diversified systems and exploring their anti-drug resistance potential. Therefore, it is particularly important to conduct a comprehensive review of the research progress of TME-targeted Nano-DDS in recent years. After a brief introduction of TME and tumor MDR, the design principle and structure of liposomes, polymer micelles and inorganic nanocarriers are focused on, and their characteristics as TME-targeted nanocarriers are described. It also demonstrates how these systems break through the cancer MDR treatment through various targeting mechanisms, discusses their synthetic innovation, research results and resistance overcoming mechanisms. The review was concluded with deliberations on the key challenges and future outlooks of targeting TME Nano-DDS in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| |
Collapse
|
41
|
Li M, Huang Y, Shen C, Wang Y, Lin Y, Wang Z, Chen N, Luo Y. Application of quantum dots in cancer diagnosis and treatment: Advances and perspectives. NANO RESEARCH 2025; 18:94907163. [DOI: 10.26599/nr.2025.94907163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
|
42
|
Amiri M, Jafari S, Lavasanifar A, Molavi O, Montazersaheb S. Nano-delivery of Silibinin Potentiate the Induction of Immunogenic Cell Death (ICD) in Melanoma Cells. Curr Pharm Biotechnol 2025; 26:392-401. [PMID: 38482616 DOI: 10.2174/0113892010280336240227062954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/17/2024] [Accepted: 02/01/2024] [Indexed: 03/25/2025]
Abstract
BACKGROUND Induction of immunogenic cell death (ICD) in tumors can enhance antitumor immunity and modulate immunosuppression in the tumor microenvironment (TME). OBJECTIVE In the current study, we investigated the effect of silibinin, a natural compound with anticancer activity, and its polymer-based nanoformulations on the induction of apoptosis and ICD in cancer cells. METHODS Free and nanoparticulate silibinin were evaluated for their growth-inhibitory effects using an MTT assay. Annexin V/PI staining was used to analyze apoptosis. Calreticulin (CRT) expression was measured by flow cytometry. Western blotting was conducted to examine the levels of elf2α, which plays a role in the ICD pathway. The HSP90 and ATP levels were determined using specific detection kits. RESULTS Compared to the free drug, silibinin-loaded nanocarriers significantly increased the induction of apoptosis and ICD in B16F10 cells. ICD induction was characterized by significantly increased levels of ICD biomarkers, including CRT, HSP90, and ATP. We also observed an increased expression of p-elf-2α/ elf-2α in B16F10 cells treated with silibinin-loaded micelles compared to cells that received free silibinin. CONCLUSION Our findings showed that the encapsulation of silibinin in polymeric nanocarriers can potentiate the effects of this drug on the induction of apoptosis and ICD in B16F10 melanoma cells.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Jafari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
43
|
Jawaid S, Joshi Y, Neelofar N, Khursheed K, Shams S, Chaudhary M, Arora M, Mahajan K, Anwar F. A Cross-talk between Nanomedicines and Cardiac Complications: Comprehensive View. Curr Pharm Des 2025; 31:741-752. [PMID: 39506444 DOI: 10.2174/0113816128347223241021111914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Cardiovascular Diseases (CVDs) are the leading cause of global morbidity and mortality, necessitating innovative approaches for both therapeutics and diagnostics. Nanoscience has emerged as a promising frontier in addressing the complexities of CVDs. OBJECTIVE This study aims to explore the interaction of CVDs and Nanomedicine (NMs), focusing on applications in therapeutics and diagnostics. OBSERVATIONS In the realm of therapeutics, nanosized drug delivery systems exhibit unique advantages, such as enhanced drug bioavailability, targeted delivery, and controlled release. NMs platform, including liposomes, nanoparticles, and carriers, allows the precise drug targeting to the affected cardiovascular tissues with minimum adverse effects and maximum therapeutic efficacy. Moreover, Nanomaterial (NM) enables the integration of multifunctional components, such as therapeutic agents and target ligands, into a single system for comprehensive CVD management. Diagnostic fronts of NMs offer innovative solutions for early detection and monitoring of CVDs. Nanoparticles and nanosensors enable highly sensitive and specific detection of Cardiac biomarkers, providing valuable insights into a disease state, its progression, therapeutic outputs, etc. Further, nano-based technology via imaging modalities offers high high-resolution imaging, aiding in the vascularization of cardiovascular structures and abnormalities. Nanotechnology-based imaging modalities offer high-resolution imaging and aid in the visualization of cardiovascular structures and abnormalities. CONCLUSION The cross-talk of CVDs and NMs holds tremendous potential for revolutionizing cardiovascular healthcare by providing targeted and efficient therapeutic interventions, as well as sensitive and early detection for the improvement of patient health if integrated with Artificial Intelligence (AI).
Collapse
Affiliation(s)
- Shagufta Jawaid
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Yogesh Joshi
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Nauroz Neelofar
- Department of Obstetrics and Gynae, Himaliyan Institute of Medical Sciences, Swami Rama Himaliyan University, Jollygrand, Dehradun, Uttarakhand, India
| | - Khuzamah Khursheed
- Shri Guru Ram Rai Institute of Medical and Health Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand, India
| | - Samya Shams
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Mansi Chaudhary
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Mitali Arora
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Karan Mahajan
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
44
|
Chowdhury D, Das A, Mishra M, Khutere T, Bodakhe SH. Physiological markers for immunotherapeutics: a review. J Chemother 2024:1-24. [PMID: 39711144 DOI: 10.1080/1120009x.2024.2443701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Immunotherapy has been advanced through multiple approaches, including immunogenic cytokines, monoclonal antibodies, therapeutic vaccinations, adoptive cell transfer, stem cell transplantation, and oncolytic viruses. This review analyses various strategies in genomics, transcriptomics, single-cell techniques, computational analysis, big data, and imaging technologies for the identification of tumour microbiota and microenvironments. Immunotherapy is becoming acknowledged as a feasible cancer treatment method, facilitating innovative cancer medicines and personalized medicine techniques.
Collapse
Affiliation(s)
- Durlav Chowdhury
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Mrityunjay Mishra
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Trinkal Khutere
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
45
|
Song Z, Chen H, Wang X, Zhang Z, Li H, Zhao H, Liu Y, Han Q, Zhang J. Napabucasin-loaded PLGA nanoparticles trigger anti-HCC immune responses by metabolic reprogramming of tumor-associated macrophages. J Transl Med 2024; 22:1125. [PMID: 39707412 DOI: 10.1186/s12967-024-05917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND JAK/STAT3 is one of the critical signaling pathways involved in the occurrence and development of hepatocellular carcinoma (HCC). BBI608 (Napabucasin), as a novel small molecule inhibitor of STAT3, has shown previously excellent anti-HCC effects in vitro and in mouse models. However, low bioavailability, high cytotoxicity and other shortcomings limit its clinical application. In this study, PLGA was selected to prepare Napabucasin PLGA nanoparticles (NPs) by solvent evaporation method, overcoming these limitations and improving the passive targeting effect that nanoparticle mediated. Base on this, we systematically evaluated the anti-HCC effect of Napabucasin-PLGA NPs and explored the underlying mechanisms. METHODS Napabucasin-PLGA NPs were prepared by solvent evaporation method. CCK-8 assay, Annexin V/PI double staining, RT-qPCR, colony formation assay, and Western blotting were performed to evaluate the anti-HCC effect of Napabucasin-PLGA NPs in vitro. Proliferation assay and migration assay were used to detect the effects of Napabucasin-PLGA NPs-treated HCC-TAMs on tumor biological characteristics of HCC cells. Flow cytometry was used to detect anti-HCC immune responses induced by Napabucasin-PLGA NPs in vivo. RESULTS Our results demonstrated that Napabucasin-PLGA NPs could improve the bioavailability of Napabucasin and enhance Napabucasin-mediated the anti-HCC effects in vitro and in vivo with no significant drug toxicity. In addition to the direct inhibitory effects on the tumor biological characteristics of HCC cells, Napabucasin-PLGA NPs could promote the polarization of macrophages from tumor-promoting M2-type to anti-tumor M1-type, improving the tumor immune microenvironment and augmenting T cell-mediated anti-tumor responses. The underlining mechanisms showed Napabucasin-PLGA NPs suppressed the STAT3/FAO signaling axis in HCC-induced tumor-associated macrophages (TAMs). CONCLUSIONS These findings demonstrated Napabucasin-PLGA NPs is a potential therapeutic candidate for HCC, and provided a new theoretical and experimental basis for further development and clinical application of Napabucasin.
Collapse
Affiliation(s)
- Zhenwei Song
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Hongfei Chen
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xueyao Wang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yang Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
46
|
Xie TQ, Yan X, Qin YT, Zhang C, Jin XK, Li QR, Rao ZY, Zhou H, Chen WH, Zhang XZ. Lactate/Cysteine Dual-Consuming Probiotic-Nanomedicine Biohybrid System for Enhanced Cancer Chemo-Immunotherapy. NANO LETTERS 2024; 24:16132-16142. [PMID: 39641324 DOI: 10.1021/acs.nanolett.4c04938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Immunotherapy is revolutionizing oncology, but its therapeutic efficiency is still limited by the off-target toxicities and poor antitumor immune responses. By integrating the drug-loaded nanoparticles (DMnSH) with the unique metabolic traits of Veillonella parvula (Vei), a probiotic-nanomedicine conjugate Vei@DMnSH biohybrid is elaborately designed for enhanced cancer chemo-immunotherapy. Specifically, Vei@DMnSH can accumulate in hypoxic tumor sites and simultaneously consume lactate and cysteine to reverse the lactate-associated immunosuppression and impede the biosynthesis of GSH. In addition, the DMnSH nanoparticles will rapidly deplete intracellular GSH and disassemble to release DOX and Mn2+. Accompanied by the two-pronged GSH depletion, the Mn2+-mediated Fenton-like reaction can effectively generate oxidative hydroxyl radicals to induce heavy redox imbalance. Combined with the therapeutic effect of DOX, robust immunogenic cell death is provoked and subsequently activates antitumor adaptive immunity with a tumor suppression rate over 82%, synergistically enhancing the therapeutic outcomes of cancer chemo-immunotherapy.
Collapse
Affiliation(s)
- Tian-Qiu Xie
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xiao Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - You-Teng Qin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xiao-Kang Jin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qian-Ru Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Zhi-Yong Rao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Hao Zhou
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan 430071, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
47
|
Ayyadurai P, Ragavendran C. Nano-bio-encapsulation of phyto-vaccines: a breakthrough in targeted cancer immunotherapy. Mol Biol Rep 2024; 52:58. [PMID: 39692899 DOI: 10.1007/s11033-024-10164-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
Nano bio-encapsulation of phyto-vaccines for cancer has marked a cutting-edge strategy that brings together nanotechnology with plant-derived vaccines to enhance cancer therapy. Phyto-vaccines, isolated from bioactive compounds found in plants called protein bodies, have been shown to potentially stimulate the immune system to recognise and destroy cancer cells. However, challenges such as poor stability, rapid degradation, and limited bioavailability in the body have hindered their clinical application. Nano bio-encapsulation offers a solution by packaging these phyto-vaccines into nanoscale carriers such as lectins have provided ways to overcome these limitations. They protect the protein bodies from degradation by proteolytic enzymes, enhance targeted delivery to cancer cells, and enable controlled release. This approach not only improves the bio-distribution and potency of the vaccines but also minimizes side effects, making it a highly promising, sustainable, and efficient method for cancer immunotherapy. As research progresses, this technology has the potential to revolutionize cancer treatment by providing safer and more precise therapeutic options. This review focuses on the concept of nano bio-encapsulation of phyto-vaccines for cancer treatment. It explores how nanotechnology can enhance the stability, bioavailability, and targeted delivery of plant-derived vaccines, addressing the limitations of traditional vaccines. The review delves into the potential of this innovative strategy to advance cancer immunotherapy, providing a comprehensive overview of current research and future directions.
Collapse
Affiliation(s)
- Pavithra Ayyadurai
- Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, India
| | - Chinnasamy Ragavendran
- Department of Cariology, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha Dental College and Hospitals, Saveetha University, Chennai, 600 077, India.
| |
Collapse
|
48
|
Chen Q, Wan W, Zhao Q, Li J, Xiong Y, Yuan Y, Tang L, Wu X, Xing W, Guo W, Lu D, Ao L, Xu X, Ao X. Decoding the prognostic landscape of LUAD: the interplay between N 6-methyladenosine modification and immune microenvironment. Front Immunol 2024; 15:1514497. [PMID: 39720723 PMCID: PMC11666524 DOI: 10.3389/fimmu.2024.1514497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Background To determine the role of N6-methyladenosine (m6A) modification in the tumor immune microenvironment (TIME), as well as their association with lung adenocarcinoma (LUAD). Methods Consensus clustering was performed to identify the subgroups with distinct immune or m6A modification patterns using profiles from TCGA. A risk score model was constructed using least absolute shrinkage and selection operator regression and validated in two independent cohorts and LUAD tissue microarrays. For experimental validation, the regulation of METTL3/m6A axis in the expression of candidate genes by RIP-qPCR assay in A549 and H460 cell lines. Co-culture experiments with human T cells were performed to evaluate the impact of METTL3 on the enhancement of anti-tumor immunity through in vitro experiments. Results We identified 282 m6A regulator genes and 955 immune-related genes, selecting seven key genes (SFTPC, CYP24A1, KRT6A, PTTG1, S100P, FAM83A, and ANLN) to develop a risk score model using Lasso regression. High-risk patients, determined by this model, exhibited poorer prognosis, increased immune infiltration, higher tumor mutational burden, more neoantigens, and elevated PD-L1 expression. These findings were validated by two independent databases and LUAD tissue microarrays. METTL3 was found to impact the mRNA expression of these genes, with METTL3 deficiency abolishing these interactions. Inhibition of METTL3 enhanced anti-tumor immunity, T cell activation, exhaustion, and infiltration in vitro. Conclusion This risk score system shows promise for prognostic prediction and the development of personalized treatment strategies for LUAD patients.
Collapse
Affiliation(s)
- Quan Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
| | - Weijun Wan
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Qing Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Juan Li
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yanli Xiong
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yuchuan Yuan
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Lu Tang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaofeng Wu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Xing
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Guo
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Di Lu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
| | - Luoquan Ao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiang Xu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Xiang Ao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Department of orthopedics, 953 Hospital of PLA Army, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, China
- Institute of Cancer, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
49
|
Fei B, Yu M, Wang Z, Li S. Biomimetic Nanovaccines Restore Immunosuppressive Tumor Antigen-Presenting Cells via the Saposin-Feeding Strategy. ACS Biomater Sci Eng 2024; 10:7482-7491. [PMID: 39560472 DOI: 10.1021/acsbiomaterials.4c01337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Cancer cell membrane-derived biomimetic nanovaccines have shown tremendous potential in cancer immunotherapy. However, their efficacy is restricted by the insufficient cross-presentation of cell membrane-associated antigens. Saposins (SAs), which are vital for membrane vesicle disintegration and cell membrane-associated antigen presentation, are severely deficient in the antigen-presenting cells (APCs) within tumors. Herein, we propose a complementary strategy for increasing the efficacy of biomimetic nanovaccines via the use of SAs. Biomimetic nanovaccines were designed using cancer cell membrane shells to provide a comprehensive array of tumor-associated antigens and reactive oxygen species (ROS)-responsive nanoparticle cores that allowed the codelivery of cytosine-guanine dinucleotides (CpGs) and SAs. The biomimetic nanovaccines were ROS-responsive and highly internalized by APCs, which enabled the release of CpGs and SAs in the endo/lysosomes of APCs. Furthermore, biomimetic nanovaccines increased the activation of immunosuppressive APCs and enhanced T-cell priming by delivering SAs to the APCs. Consequently, biomimetic nanovaccines loaded with SAs not only suppressed tumor growth but also exhibited excellent therapeutic effects in combination with immune checkpoint blockade strategies. Overall, our study provides insights into the development of enhanced biomimetic nanovaccines via integrating SAs and offers a promising strategy for highly effective cancer immunotherapy.
Collapse
Affiliation(s)
- Bingyuan Fei
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Miao Yu
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China
| | - Shuo Li
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| |
Collapse
|
50
|
Lucas S, Thomas SN. Therapeutic Immunomodulation of Tumor-Lymphatic Crosstalk via Intratumoral Immunotherapy. Mol Pharm 2024; 21:5929-5943. [PMID: 39478434 PMCID: PMC11615947 DOI: 10.1021/acs.molpharmaceut.4c00692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024]
Abstract
Intra- and peritumoral lymphatics and tumor-draining lymph nodes play major roles in mediating the adaptive immune response to cancer immunotherapy. Despite this, current paradigms of clinical cancer management seldom seek to therapeutically modulate tumor-lymphatic immune crosstalk. This review explores recent developments that set the stage for how this regulatory axis can be therapeutically manipulated, with a particular emphasis on tumor-localized immunomodulation. Building on this idea, the nature of tumor-lymphatic immune crosstalk and relevant immunotherapeutic targets and pathways are reviewed, with a focus on their translational potential. Engineered drug delivery systems that enhance intratumoral immunotherapy by improving drug delivery to both the tumor and lymph nodes are also highlighted.
Collapse
Affiliation(s)
- Samuel
N. Lucas
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
of America
| | - Susan N. Thomas
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
of America
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States of America
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States of America
- Winship
Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
of America
| |
Collapse
|